Download as pdf or txt
Download as pdf or txt
You are on page 1of 453

Dennis M.

Styne

Pediatric
Endocrinology

A Clinical Handbook

123
Pediatric Endocrinology
Dennis M. Styne

Pediatric Endocrinology
A Clinical Handbook
Dennis M. Styne
Pediatrics
University of California
Davis School of Medicine
Sacramento, CA, USA

ISBN 978-3-319-18370-1 ISBN 978-3-319-18371-8 (eBook)


DOI 10.1007/978-3-319-18371-8

Library of Congress Control Number: 2015940868

© Springer International Publishing Switzerland 2016


This work is subject to copyright. All rights are reserved by the Publisher, whether the whole or
part of the material is concerned, specifically the rights of translation, reprinting, reuse of
illustrations, recitation, broadcasting, reproduction on microfilms or in any other physical way,
and transmission or information storage and retrieval, electronic adaptation, computer software,
or by similar or dissimilar methodology now known or hereafter developed.
The use of general descriptive names, registered names, trademarks, service marks, etc. in this
publication does not imply, even in the absence of a specific statement, that such names are
exempt from the relevant protective laws and regulations and therefore free for general use.
The publisher, the authors and the editors are safe to assume that the advice and information in
this book are believed to be true and accurate at the date of publication. Neither the publisher nor
the authors or the editors give a warranty, express or implied, with respect to the material
contained herein or for any errors or omissions that may have been made.

Printed on acid-free paper

This Springer imprint is published by Springer Nature


The registered company is Springer International Publishing AG Switzerland
To my loving wife Donna and my amazing children
Rachel, Jonathan, Juliana, and Aaron, my wonderful
son-in-law Michael, and my beautiful grandchildren
Cooper and Samara all of whom immeasurably enrich my life.
Preface

This book is written for practitioners ranging from students to residents, fam-
ily physicians, and pediatricians puzzling over the approach to a child who
seems to have an endocrine disorder in order to help readers evaluate and
treat the more straightforward pediatric endocrine issues. The information in
the text might prepare a provider for a meeting with parents of a child with an
endocrine condition in order to improve communication or alternatively
might provide the facts that will allow the practitioner to avoid an unneces-
sary consultation. The chapters begin with the basic physiologic processes
and concepts of endocrinology before putting this knowledge into action by
addressing the major signs and symptoms and the diagnosis and management
of pediatric endocrine disorders. This work is provided for anyone who
wishes to have a consultation with a pediatric endocrinologist in absentia still
realizing that an actual person-to-person consultation sets the gold standard.
There are a multitude of more complex issues within pediatric endocrinology
that cannot be mastered without adequate experience; managing a baby with
ambiguous genitalia is only one example of the issues that are introduced in
this text to allow the reader to understand the basics, but this small volume
cannot possibly cover the complexities in their entirety. This volume is not
meant to replace the outstanding larger and more detailed texts, manuscripts,
and websites to which I refer the reader in the “suggested reading” section at
the conclusion of each chapter. The concepts covered are designed to address
in varying detail the subjects in the first ten headings in the content outline of
the Sub-board of Pediatric Endocrinology of The American Board of
Pediatrics dealing with clinical conditions. However, there is no claim that
this book will give the reader the expertise of a pediatric endocrinologist; for
that a 3-year fellowship is only the first step!

Sacramento, CA, USA Dennis M. Styne

vii
Acknowledgements

I am indebted to my colleagues in the field of pediatric endocrinology across


the world whose wisdom in print and in words I value and have tried to reflect
in this book. This text grew out of my teaching and clinical experiences and
I am grateful to the innumerable students, house officers, and postgraduate
practitioners I have encountered in the last 45 years that I have pursued this
field. Presentations to them helped me organize my thoughts and their probing
questions led me to look at the issues in new ways. I am grateful to my first
teachers of this field, Melvin M. Grumbach, Felix Conte, and the late Selna
Kaplan, at the University of California San Francisco. They deserve much of
the credit for my accomplishments as they started me in this wonderful field.
I am indebted to my coworkers at the University of California Davis School
of Medicine, Drs. Nicole Glaser, Lindsey Albrecht, Abigail Fruzza, and
Yvonne Lee: they have daily imparted their wisdom to me. My clinical team
that provide outstanding care every day, especially to our numerous patients
with diabetes mellitus, include Sultanna Iden, RN, CDE; Erin Conboy-Heiser,
RN, CDE; Alexander Nella, RD, CDE; Dayna Green-Burgeson, RD, CDE;
Vincent Fong, LCSW; and Breanne Harris, BS Diabetes Concierge. Edna
Gun Utter, our AA (administrative assistant), helped me organize and for-
mat this volume expertly. The supportive staff at Springer including Richard
Lansing and Joseph Quatela gave indispensable editorial help. I acknowledge
Dr. Edward Steinberg who provided initial support in my redirection into
pediatric obesity and continues to provide constant wisdom. I am deeply
indebted to the Yocha Dehe Wintun Nation who have endowed my faculty
position and unwaveringly supported my work in pediatric endocrinology
and health disparities over the last 15 years.

ix
Contents

1 Introduction to Pediatric Endocrinology:


The Endocrine System .................................................................. 1
Suggested Readings ........................................................................ 10
2 The Evaluation of a Child or Adolescent
with Possible Endocrine Disease .................................................. 11
The Medical History ....................................................................... 11
Physical Examination...................................................................... 13
3 Disorders of the Hypothalamic–Pituitary Axis .......................... 17
Physiology....................................................................................... 17
Pathology ........................................................................................ 19
Central Nervous System Tumors .................................................... 19
Craniopharyngioma..................................................................... 20
Germinomas ................................................................................ 21
Pituitary Adenomas......................................................................... 22
Other Central Nervous System Disorders ....................................... 23
Langerhans Cell Histiocytosis .................................................... 23
Postinfectious Inflammatory Lesions of the Central
Nervous System, Vascular Abnormalities,
and Head Trauma ........................................................................ 23
Irradiation of the Central Nervous System ..................................... 24
Developmental Defects of the Midline ........................................... 24
Septo-Optic and Optic Dysplasia ................................................ 24
The Solitary Median Maxillary Incisor Syndrome ..................... 25
Idiopathic Hypopituitary Dwarfism ................................................ 25
Suggested Readings ........................................................................ 27
4 The Posterior Pituitary Gland and Disorders
of Vasopressin Metabolism ........................................................... 29
Normal Vasopressin Physiology ..................................................... 29
Diabetes Insipidus ........................................................................... 33
Central Diabetes Insipidus .......................................................... 33
Nephrogenic Diabetes Insipidus ................................................. 35
Clinical Features of Diabetes Insipidus ...................................... 36

xi
xii Contents

Diagnosis of Diabetes Insipidus.................................................. 36


Treatment of Central Diabetes Insipidus .................................... 39
Treatment of Nephrogenic Diabetes Insipidus............................ 41
The Syndrome of Inappropriate Secretion
of Antidiuretic Hormone ................................................................. 41
Nephrogenic Syndrome of Inappropriate Antidiuresis ................... 42
Other Causes of Hyponatremia ....................................................... 43
The Treatment of Syndrome of Inappropriate
Secretion of Antidiuretic Hormone ................................................. 43
The Triphasic Response After Surgery
for Craniopharyngioma ................................................................... 44
Suggested Readings ........................................................................ 45
5 Disorders of Growth ..................................................................... 47
Measurement of Growth ................................................................. 47
Endocrine Factors in Postnatal Growth .......................................... 58
Growth Hormone ........................................................................ 58
Insulin-Like Growth Factor or IGF ............................................. 62
Epidermal Growth Factor............................................................ 66
Erythropoietin ............................................................................. 66
Oncogenes ................................................................................... 66
Dental Development ....................................................................... 67
Abnormalities of Growth ................................................................ 67
Short Stature................................................................................ 67
Growth and Attention-Deficit Disorder ...................................... 73
Tall Stature .................................................................................. 86
Suggested Readings ........................................................................ 89
6 Disorders of the Thyroid Gland ................................................... 91
Normal Thyroid Physiology and Anatomy ..................................... 91
Fetal Thyroid Development in the Newborn .................................. 95
Laboratory Evaluation..................................................................... 97
Imaging of Thyroid ......................................................................... 98
Disorders of the Thyroid Gland ...................................................... 99
Goiter .......................................................................................... 99
Hypothyroidism .............................................................................. 101
Congenital Hypothyroidism (CH)............................................... 101
Acquired Hypothyroidism .......................................................... 108
Thyroid Hormone Overdose ....................................................... 117
Neoplasms ................................................................................... 117
Painful Thyroid Glands ............................................................... 120
Further Reading .............................................................................. 120
7 Disorders of Calcium Metabolism and Bone .............................. 123
Calcium Metabolism ....................................................................... 123
Storage and Absorption of Calcium............................................ 123
The Calcium-Sensing Receptor .................................................. 124
Phosphate .................................................................................... 124
Magnesium.................................................................................. 125
Parathyroid Hormone and Parathyroid
Hormone-Related Protein ........................................................... 126
Contents xiii

Vitamin D and Its Metabolites .................................................... 127


Calcitonin .................................................................................... 129
Alkaline Phosphatase .................................................................. 129
Hypocalcemia ................................................................................. 130
Hypocalcemia in the Infant ......................................................... 130
Hypocalcemia in the Child and Adolescent ................................ 133
Diagnosis of Hypocalcemia ........................................................ 139
Normal Bone Growth .................................................................. 139
Vitamin D and Calcium Deficiency ............................................ 140
Rickets......................................................................................... 141
Hypercalcemia ................................................................................ 147
Primary Hyperparathyroidism .................................................... 149
Familial Hypocalciuric Hypercalcemia ...................................... 149
Miscellaneous Causes ................................................................. 150
Treatment of Hypercalcemia ....................................................... 152
Evaluation of Hypercalcemia ...................................................... 152
Aluminum Toxicity ..................................................................... 156
Suggested Readings ........................................................................ 157
8 Disorders of Sexual Differentiation ............................................. 159
Normal Sexual Differentiation ........................................................ 159
Genotype ..................................................................................... 159
Gonadal Sex ................................................................................ 161
Phenotypic Sex............................................................................ 162
Social Gender .............................................................................. 165
Disorders of Sexual Differentiation ................................................ 166
46, XX DSD: Conditions Causing Genetic
Females to Become Virilized (Previously Called
Female Pseudohermaphroditism)................................................ 166
46, XY DSD: Disorders Causing Inadequate
Virilization of a Genetic Male (Previously Called
Male Pseudohermaphroditism) ................................................... 175
Gonadal DSD .............................................................................. 179
Endocrine Disruptors .................................................................. 180
The Diagnosis and Treatment of Ambiguous Genitalia.................. 180
Treatment ........................................................................................ 184
The Decision of Sex of Rearing ...................................................... 184
Multidisciplinary Team Management ............................................. 186
Youth with Gender Dysphoria .................................................... 186
Suggested Readings ........................................................................ 186
9 Disorders of Puberty ..................................................................... 189
Normal Pubertal Development........................................................ 189
Physical Development................................................................. 189
Age at Onset of Puberty .............................................................. 191
Ages of Pubertal Stages and Duration ........................................ 193
Skeletal Development and Bone Mineral Density ...................... 193
The Endocrine Changes of Puberty ................................................ 193
Gonadarche ................................................................................. 195
xiv Contents

Spermarche ................................................................................. 200


Adrenarche .................................................................................. 200
Pediatric Sex Steroid and Gonadotropin\ Assays ........................ 200
Ovulation and Menarche ............................................................. 201
Miscellaneous Metabolic Changes ............................................. 202
Abnormalities of Puberty ................................................................ 202
Delayed Puberty .......................................................................... 202
Sexual Precocity.......................................................................... 217
Endocrine Disruption in Puberty .................................................... 224
Variations of Early Pubertal Development.................................. 224
Macroorchidism .......................................................................... 225
Differential Diagnosis of Precocious Puberty............................. 225
Treatment of Precocious Puberty ................................................ 226
Polycystic Ovarian Syndrome ......................................................... 229
Suggested Readings ........................................................................ 230
10 Disorders of the Adrenal Gland................................................... 233
Normal Adrenal Gland Physiology ................................................. 233
Measurement of Steroids ................................................................ 239
Disorders of the Adrenal Cortex ..................................................... 240
Hypoadrenal States ..................................................................... 240
Aldosterone Insufficiency or Resistance ..................................... 248
Adrenal Calcifications................................................................. 248
Hyperadrenal States .................................................................... 249
Hypertension due to Adrenal Disease ......................................... 256
Adrenal Medulla ............................................................................. 257
Normal Physiology ..................................................................... 257
Disorders of the Adrenal Medulla............................................... 258
Suggested Reading .......................................................................... 261
11 Diabetes Mellitus ........................................................................... 263
Insulin Biosynthesis and Action ..................................................... 265
Glucagon ......................................................................................... 266
Other GI Hormones......................................................................... 267
Diabetes Mellitus ............................................................................ 270
Type 1 Diabetes Mellitus ................................................................ 270
Clinical Presentation of Diabetes Mellitus ................................. 272
Dietary Management ................................................................... 280
Preparation for Discharge ........................................................... 280
Home Measurement by Glucometer ........................................... 281
Subcutaneous Insulin Management ............................................ 282
Glycosylated Hemoglobin or Hemoglobin A1c ........................... 287
The Honeymoon Period .............................................................. 288
Sick Day Management ................................................................ 288
Hypoglycemia ............................................................................. 289
Somogyi Phenomenon ................................................................ 290
Dawn Phenomenon ..................................................................... 291
Surgery and Diabetes .................................................................. 291
Contents xv

Diabetes Team Management ........................................................... 291


Iatrogenic or Other Acquired Forms
of Diabetes Mellitus Type 1 ........................................................ 293
Neonatal Diabetes ........................................................................... 297
Transient Neonatal Diabetes Mellitus ......................................... 297
Permanent Neonatal Diabetes Mellitus ....................................... 298
Type 2 Diabetes Mellitus ................................................................ 298
Wolfram Syndrome ..................................................................... 301
Maturity-Onset Diabetes of the Young
or Monogenic Diabetes ............................................................... 301
Mitochondrial Disease and Diabetes .......................................... 301
Nonketotic Hyperosmotic Coma................................................. 302
Summary ......................................................................................... 303
Suggested Readings ........................................................................ 303
12 Hypoglycemia ................................................................................ 305
Normal Carbohydrate Metabolism ................................................. 305
Glucose Regulatory Factors ........................................................ 306
Transition of the Fetus to Neonatal Life ..................................... 309
Hypoglycemia ................................................................................. 309
Hypoglycemia in the Newborn ................................................... 310
Hypoglycemia in Older Children ................................................ 316
Diagnosis of Hypoglycemia........................................................ 319
Treatment of Hypoglycemia ....................................................... 322
Suggested Reading .......................................................................... 324
13 Obesity ........................................................................................... 325
Definition ........................................................................................ 325
Etiology of Obesity ......................................................................... 329
Genetic Factors and the CNS Control of Appetite...................... 329
Dietary Intake.............................................................................. 331
Energy Expenditure..................................................................... 336
Gut Microbiome .......................................................................... 336
Epigenetics .................................................................................. 336
The Obesogenic Environment ..................................................... 337
Endocrine Causes ........................................................................ 338
Iatrogenic Causes ........................................................................ 339
Comorbidities of Childhood and Adolescent Obesity .................... 339
Psychological Conditions............................................................ 340
Type 2 Diabetes........................................................................... 340
The Metabolic Syndrome and Insulin Resistance ....................... 340
Polycystic Ovarian Syndrome ..................................................... 342
Hyperlipidemia and Risk for Cardiovascular Disease ................ 342
Hypertension ............................................................................... 347
Liver Disease............................................................................... 349
Obstructive Sleep Apnea ............................................................. 350
Dermal Findings.......................................................................... 351
Orthopedic Conditions ................................................................ 351
Increased Growth and Advanced Skeletal Development ............ 351
xvi Contents

Earlier Puberty ............................................................................ 351


Enuresis ....................................................................................... 351
Cholelithiasis............................................................................... 351
Asthma ........................................................................................ 351
Pseudotumor Cerebri .................................................................. 351
Psoriasis ...................................................................................... 352
Laboratory Evaluation of Childhood Obesity ................................. 352
Treatment .................................................................................... 352
Lifestyle Modification................................................................. 353
Medication .................................................................................. 355
Surgery ........................................................................................ 356
Prevention ................................................................................... 357
Encourage Parental Modeling ..................................................... 357
Expose Children to Healthy Food in a Young Age ..................... 357
Encourage Breast-Feeding .......................................................... 357
Decrease or Eliminate the Intake of Sugar Sweetened
Beverages and Limit the Intake of Juice ..................................... 358
Increase Public Safety ................................................................. 358
Modify the Diet ........................................................................... 358
Decrease Sedentary Time............................................................ 358
Suggested Readings ........................................................................ 360
14 Pediatric Endocrine Emergencies................................................ 363
Acute Adrenal Insufficiency ........................................................... 363
Diabetic Ketoacidosis ..................................................................... 365
Hypocalcemia ................................................................................. 366
Hypercalcemia ................................................................................ 368
Hyperthyroidism in the Neonate ..................................................... 370
Thyroid Storm in Children and Adolescents .................................. 370
Hypoglycemia ................................................................................. 371
Hyponatremia/Hyperkalemia Due to Adrenal Insufficiency .......... 373
Hyponatremia Resulting from Syndrome
of Inappropriate Secretion of Antidiuretic Hormone ...................... 374
Hypernatremia Due to Diabetes Insipidus ...................................... 374
Hypertension Due to Pheochromocytoma ...................................... 375
Suggested Reading .......................................................................... 375
15 Medications for Pediatric Endocrinology ................................... 377
Calcitonin, Human .......................................................................... 377
Calcitriol (1,25-Dihydroxycholecalciferol) .................................... 377
Calcium ........................................................................................... 377
Calcium Carbonate, Oral ............................................................ 377
Calcium Citrate, Oral .................................................................. 378
Calcium Glubionate, Oral ........................................................... 378
Calcium Gluconate IV ................................................................ 378
Hypocalcemia ............................................................................. 378
Calcium Lactate, Oral ................................................................. 378
DDAVP ........................................................................................... 378
Ergocalciferol (Vitamin D2) ........................................................... 378
Dietary Supplementation ............................................................ 379
Vitamin D Deficiency.................................................................. 379
Contents xvii

Nutritional Rickets ...................................................................... 379


Vitamin D-Resistant Rickets ....................................................... 379
Vitamin D-Dependent Rickets .................................................... 379
Hypoparathyroidism or Pseudohypoparathyroidism .................. 379
Estrogen ...................................................................................... 379
Fludrocortisone Acetate (Florinef) ................................................. 379
Glucagon HCl ............................................................................. 379
Glucocorticoid Preparations............................................................ 379
Growth Hormone Secretagogue Testing ......................................... 379
Hydrochlorothiazide ....................................................................... 380
Insulin Preparations ........................................................................ 380
Thyroxine (Levo) ............................................................................ 380
Magnesium Sulfate ......................................................................... 380
Hypomagnesemia or Hypocalcemia ........................................... 380
Daily Maintenance ...................................................................... 381
Mannitol (5, 10, 15, 20, 25 %) ........................................................ 381
Medroxyprogesterone Acetate .................................................... 381
Metformin ....................................................................................... 381
Mecasermin (rhIGF-1) .................................................................... 381
Methimazole ................................................................................... 381
Hyperthyroidism ......................................................................... 381
Nitroprusside ............................................................................... 381
Pamidronate .................................................................................... 381
Hypercalcemia ............................................................................ 381
Osteogenesis Imperfecta ............................................................. 382
Phosphate Supplements .................................................................. 382
Maintenance/Replacement .......................................................... 382
Potassium Iodide ............................................................................. 382
Potassium Supplements .................................................................. 382
Hypokalemia ............................................................................... 382
Monitor Serum K Closely and Watch EKG at All Times ............... 382
Propranolol...................................................................................... 382
Hypertension ............................................................................... 382
Sodium Bicarbonate ........................................................................ 383
Somatropin (Growth Hormone) ...................................................... 383
Growth Hormone Deficiency ...................................................... 383
Noonan’s Syndrome .................................................................... 383
Prader-Willi Syndrome ............................................................... 383
SGA............................................................................................. 383
SHOX Deficiency........................................................................ 383
Turner Syndrome ........................................................................ 383
Spironolactone ................................................................................ 383
Diuretic ....................................................................................... 383
Primary Aldosteronism ............................................................... 383
Hirsutism or Acne in Women...................................................... 383
Testosterone .................................................................................... 384
Vasopressin ..................................................................................... 384
Diabetes Insipidus ....................................................................... 384
Subcutaneous/Intramuscular ....................................................... 384
Suggested Reading .......................................................................... 384
xviii Contents

16 Laboratory Values for Pediatric Endocrinology ........................ 385


Adrenocorticotropic Hormone (ACTH) Quest ............................... 385
Adrenal Antibody Screen with Reflex Titer QUEST .................. 386
Albumin (Microalbumin), 24-h Urine Quest .................................. 386
Albumin Serum Quest..................................................................... 386
Aldosterone, 24-h Urine Quest ....................................................... 386
Aldosterone Serum, LC/MS/MS Quest .......................................... 387
Alkaline Phosphatase, Serum Quest ............................................... 387
Alkaline Phosphatase, Bone-Specific Labcorp ............................... 387
Androstenedione LC/MS/MS Quest ............................................... 388
Angiotensin II Quest ....................................................................... 388
Anti-Mullerian Hormone (AMH) Quest ......................................... 389
Arginine Vasopressin Quest ............................................................ 389
Congenital Adrenal Hyperplasia 21-Hydroxylase
Deficiency Common and Rare Mutations Quest ............................ 389
C Peptide, Serum Quest .................................................................. 389
Calcitonin Quest.............................................................................. 390
Calcium, 24-h Urine Quest ............................................................. 390
Calcium Ionized, Serum Quest ....................................................... 390
Calcium, Serum Quest .................................................................... 391
Carnitine, Serum Quest ................................................................... 391
Carotene Quest ................................................................................ 391
Catecholamines, Fractionated, 24-h Urine Quest ........................... 391
Catecholamines, Fractionated, Plasma Quest ................................. 392
Catecholamines, Fractionated, Random Urine Quest ..................... 393
Corticosterone LC/MS/MS Quest ................................................... 394
Corticotropin-Releasing Hormone (Mainly Use
for Ectopic Production of CRH) Quest ........................................... 395
Cortisol-Binding Globulin (Transcortin) Quest .............................. 395
Cortisol Urinary Free 24-h Quest ................................................... 395
Cortisol-Free Serum Quest ............................................................. 395
Cortisol, Salivary......................................................................... 396
Cortisol Serum Total LC/MS/MS Esoterix ..................................... 396
Creatinine 24-h Urine Quest ........................................................... 397
Creatinine Random Urine Quest ..................................................... 397
Creatinine Clearance Quest............................................................. 397
Creatinine Serum Quest .................................................................. 398
DHEA Serum Quest Esoterix ......................................................... 398
DHEA Sulfate, Serum Esoterix ...................................................... 399
Deoxycorticosterone (DOC), Serum Esoterix ................................ 399
11-Deoxycortisol (Compound S) Corticosterone,
Serum Esoterix ................................................................................ 400
Dihydrotestosterone Esoterix .......................................................... 400
Dihydrotestosterone, Free, Serum Quest ........................................ 401
Estradiol, Serum Esoterix ............................................................... 401
Estrone, Serum Esoterix.................................................................. 402
Follicle-Stimulating Hormone, Third Generation,
Serum Esoterix ................................................................................ 402
Contents xix

Fructosamine, Serum Quest ............................................................ 403


Gastrin Serum Quest ....................................................................... 403
Glucagon Quest............................................................................... 404
Glucose, Serum ............................................................................... 404
Glutamic Acid Decarboxylase-65 Autoantibodies Quest ............... 404
Glycated Albumin Quest................................................................. 404
1,5-Anhydroglucitol Esoterix ..................................................... 405
Growth Hormone, Serum Esoterix.................................................. 405
Growth Hormone Antibody Quest .................................................. 405
Growth Hormone-Binding Protein (GHBP) Esoterix ..................... 405
Growth Hormone-Releasing Hormone Quest ................................. 406
Hemoglobin A1C, Blood Quest ........................................................ 406
Homovanillic Acid, 24-h Urine Quest ............................................ 406
17-Hydroxycorticosteroids, 24-h Urine Quest................................ 407
18-Hydroxycorticosterone, Serum Esoterix.................................... 407
5-Hydroxyindoleacetic Acid (5-HIAA), 24-h
Urine Quest ..................................................................................... 408
5-Hydroxyindoleacetic Acid (5-HIAA),
Random Urine Quest....................................................................... 408
17-Hydroxypregnenolone, Serum Esoterix .................................... 408
17-Hydroxyprogesterone (17OHP), Serum Esoterix ...................... 409
Insulin-Like Growth Factor 1 (IGF-1) (Previously
Called Somatomedin C) Esoterix.................................................... 410
IGF-2 Esoterix ................................................................................ 411
IGF-Binding Protein-1 (IGFBP-1) Quest ....................................... 411
IGF-Binding Protein-2 (IGFBP-2) Quest ....................................... 411
IGF-Binding Protein-3 (IGFBP-3) Esoterix ................................... 412
Inhibin B Quest ........................................................................... 412
Insulin Antibodies, Highly Sensitive Quest .................................... 413
Insulin, Free Quest .......................................................................... 413
Insulin, Serum Quest....................................................................... 413
Islet Cell Antibody Quest................................................................ 413
Leptin, Serum Quest ....................................................................... 413
Luteinizing Hormone (LH), Third-Generation
Serum Esoterix ................................................................................ 414
Metanephrine and VMA, Fractionated 24-h Urine Quest .............. 414
Metanephrine, Fractionated Random Urine Quest ......................... 415
Metanephrine, Fractionate Free Plasma Quest ............................... 416
Microalbumin .................................................................................. 416
Osmolality, Random Urine Quest ................................................... 416
Osmolality, Serum Quest ................................................................ 416
Osteocalcin, Serum Quest ............................................................... 416
Pancreatic Polypeptide Quest ..................................................... 417
Parathyroid Hormone Antibody, Serum Quest ............................... 417
Parathyroid Hormone (PTH), Intact, Serum Quest ......................... 417
Parathyroid Hormone and Related Protein (PTH-RP) Quest.......... 418
Pregnenolone, Serum Esoterix ........................................................ 418
xx Contents

Progesterone, Serum Esoterix ......................................................... 418


Progesterone, Free, HPLC-MS/MS ................................................ 419
Proinsulin, Serum Quest ................................................................. 419
Prolactin, Serum Quest ................................................................... 420
Renin Activity (PRA), Plasma Esoterix.......................................... 420
Sex Hormone-Binding Globulin (SHBG) Esoterix ........................ 420
Somatostatin Quest ......................................................................... 421
T3 (Triiodothyronine), Free, Nondialysis Quest.............................. 421
T3 (Triiodothyronine), Free, Dialysis Esoterix................................ 422
T3 (Triiodothyronine), Reverse Esoterix ......................................... 422
T3 (Triiodothyronine), Total, Radioimmunoassay Esoterix ............ 422
T4 (Thyroxine), Free, Direct Dialysis Serum Quest........................ 423
T4 (Thyroxine), Free, Nondialysis, Serum Esoterix........................ 423
T4 (Thyroxine), Total, Serum Quest ................................................ 424
Testosterone, Total, Serum Esoterix ............................................... 424
Testosterone Free, Serum or Plasma Esoterix................................. 425
Testosterone Saliva Esoterix ....................................................... 426
Testosterone Bioavailable Esoterix ................................................. 426
Thyroglobulin Antibody, Serum Quest ........................................... 427
Thyroid Peroxidase Antibody (Anti-TPO), Serum Quest............... 427
Thyroid-Stimulating Hormone (TSH), Serum Quest...................... 427
Thyroid-Stimulating Immunoglobulin (TSI) Quest ........................ 428
TSH Receptor Antibody (TRAb) Esoterix. .................................... 428
Thyroxine-Binding Protein/Globulin (TBG), Serum Quest ........... 428
Vanillylmandelic Acid (VMA), Random Urine Quest ................... 429
Vanillylmandelic Acid (VMA), 24-h Urine Quest .......................... 429
Vitamin D, 1,25-Dihydroxy, Serum Esoterix.................................. 429
Vitamin D, 25-Hydroxy, Serum or Plasma Esoterix ....................... 430
S.I. Unit Conversion Table from Esoterix/Labcorp ........................ 431
Suggested Reading .......................................................................... 434
Laboratory values from ............................................................... 434

Index ....................................................................................................... 435


Introduction to Pediatric
Endocrinology: The Endocrine 1
System

The endocrine system regulates reproduction, that a higher level of control of these functions
growth and development, homeostasis of the exists. Now it is recognized that various regions
organism (or maintenance of the internal envi- of the brain regulate the hypothalamus with
ronment), and the production, storage, and utili- complex interconnections in the central nervous
zation of energy. The endocrine system was system. Many of the hormones of the pituitary–
originally understood to regulate metabolism by hypothalamic axis or molecules that share much
biochemical messengers or hormones that were of their structure and function are also found in
released from specialized organs (glands) into the gastrointestinal tract and other tissues
the general circulation so that they could act at a throughout the body, as well as the placenta.
distance. Thus, hormones were classically Overlap occurs between the control of the endo-
defined as circulating messengers, with the loca- crine system and the nervous system; hormone
tion of their action far from the site of secretion, secretion can be regulated by nerve cells, and
endocrine action. Hormone action also may be endocrine agents can serve as neural messengers.
paracrine (acting on adjacent neighboring cells Further, the endocrine system is regulated by fac-
to the cell of origin of the hormone by diffusion), tors important in the immune system (e.g., cyto-
autocrine (acting on the cell of origin of the hor- kines interact with the secretion of glucocorticoids,
mone itself by diffusion), or intracrine (acting on which then exert an influence on inflammation).
the cell of origin without actually being secreted); An entire system of regulation, rather than the
often agents acting in these ways are called fac- simplistic view of the endocrine system, best
tors rather than hormones. Indeed, these factors describes the control of metabolism.
(e.g., growth factors) may be produced in most Many hormones are regulated in a feedback
cells of the body rather than discrete endocrine loop, so that the production of a hormone is
glands (Fig. 1.1). The effects of hormones and controlled by its effect; for example, the
factors may be generally considered to be corticotropin-releasing factor (CRF) stimulates
directed toward the processes of cell differentia- the adrenocorticotropic hormone (ACTH) to
tion, cell growth, and metabolism of the cell and produce cortisol, which in turn feeds back to
the organism. suppress CRF and ACTH production so that
In the past, some of the endocrine glands equilibrium is reached and serum cortisol and
appeared to be controlled by the pituitary gland, ACTH remain in the normal range (Fig. 1.2,
which was therefore considered to be “the master Table 1.1). The set point of the equilibrium may
gland.” The discovery of the hypophysiotropic change with development; in prepuberty, small
hormones of the hypothalamus and their role in amounts of sex steroids strongly suppress
the control of pituitary secretion made it clear gonadotropin secretion, but during pubertal

© Springer International Publishing Switzerland 2016 1


D.M. Styne, Pediatric Endocrinology, DOI 10.1007/978-3-319-18371-8_1
2 1 Introduction to Pediatric Endocrinology: The Endocrine System

Fig. 1.1 Endocrine, paracrine, autocrine, and intracrine cell of origin, in this case via a peptide cellular membrane
effects. Endocrine effects occur when a hormone is receptor, without entering the bloodstream nor necessarily
secreted by a cell into the bloodstream to be carried a dis- affecting nearby cells. Intracrine effects occur when a
tance until it leaves the bloodstream to interact with the substance acts within the cell in which it was produced
cellular receptor, in this case a peptide cellular membrane without crossing the cell membrane, in this case via an
receptor, to exert intracellular activity. Paracrine effects intracellular receptor. Endocrine effects are measured by
occur when a cell secretes a substance to cause biologi- assays on blood samples, but paracrine and autocrine
cal effects on a neighboring cell, in this case via a pep- effects cannot be directly measured by assaying blood
tide cellular membrane receptor, without entering the samples. In fact, the results of blood sample analyses only
bloodstream. Autocrine effects occur when the cell provide a reflection of what might be going on locally
secretes a substance to cause biological effects on the

development, the sensitivity of this feedback loop without considering its controlling factors or the
decreases. Thus increased sex steroid production effects it exerts.
that brings about the ensuing physical changes of Endocrine disorders may manifest in several
puberty occurs because these pubertal levels of ways:
sex steroids can no longer suppress gonadotropin
secretion, as would occur at a similar level of sex 1. By excess hormone effect (e.g., in Cushing syn-
steroid secretion in the prepubertal state. drome, an excess of glucocorticoid is present;
A clinician may deduce the level of an endo- e.g., if the excess is secondary to autonomous
crine defect in the system by measuring the con- glucocorticoid secretion by a target organ (cor-
centrations of hormones in serum or plasma at tisol secretion by the adrenal gland), the trophic
various steps of the process (e.g., a low serum hormone ACTH will be suppressed).
cortisol and high adrenocortical-stimulating hor- 2. By deficient hormone: e.g., in glucocorticoid
mone (ACTH) indicates a primary defect at the deficiency, inadequate cortisol is present; if
level of the adrenal gland, whereas a low serum the deficiency is at the target organ (the adre-
cortisol along with a low ACTH indicates a disor- nal gland), the trophic hormone (ACTH) will
der of the pituitary gland or the hypothalamus be elevated.
(Table 1.1)). Positive feedback loops occur as 3. By an abnormal response of end organ to a
well; the midcycle LH peak which triggers ovula- hormone: e.g., in pseudohypoparathyroidism,
tion occurs due to positive estrogen feedback on resistance to parathyroid hormone (PTH)
higher CNS centers. In most cases, it is not suf- occurs, and so PTH is elevated, but the para-
ficient to obtain a single hormone measurement thyroid hormone exerts no effects.
1 Introduction to Pediatric Endocrinology: The Endocrine System 3

a Third ventricle Hypothalamus


Neuroendocrine
cell nuclei

CRF secreting cells

Superior
hypophyseal
artery
Stalk

Long portal
vessels
Inferior
Adrenocorticotrophs hypophyseal
artery

Secreting cells

Anterior Posterior
pituitary pituitary

Hormone Short portal


secretion vessel
Glomerulosa
ACTH
Fasciculata

Reticularis

Medulla

Aldosterone Cortisol Androgens Catecholamines

Fig. 1.2 Logic of evaluation of hypothalamic–pituitary increases due to absence of negative feedback inhibition.
feedback loops using the hypothalamic–pituitary–adrenal (c) Decreased or “hypo” situation whereby the stimula-
axis as an example. (a) The normal situation where the tory hormone is absent and the target gland’s hormone
target gland (adrenal gland) regulates the stimulatory hor- product falls. In the hypothalamic–pituitary–target gland
mone by negative feedback inhibition. (b) Primary defect axis, this would be a secondary (ACTH secretory defect
due to failure of the target gland and decrease in its secre- illustrated here) or tertiary (CRF secretory defect not
tion of hormone: the stimulatory hormone (ACTH) illustrated in the figure) defect

4. By gland enlargement that may cause effects as production of ACTH from an oat cell carcinoma
a result of size rather than function; with a large of the lung in adults. Endocrine disorders may be
nonfunctioning pituitary adenoma, abnormal revealed by the response of various organs to an
visual fields and other neurological signs and excess or deficiency of various hormones well
symptoms will result, and pituitary endocrine before the size of the endocrine tumor makes it
cells may cease to function, even though no apparent (adrenal carcinoma may cause viriliza-
hormone is produced by the tumor itself. tion before it exerts a mass effect).
Hormones have various molecular structures.
Tumors of other nonendocrine organs also
Thus there are peptide (e.g., TRF), glycoprotein
may produce hormones as with the ectopic
(LH), monoamine (epinephrine), amino acid
4 1 Introduction to Pediatric Endocrinology: The Endocrine System

b Third ventricle Hypothalamus


Neuroendocrine
cell nuclei

CRF secreting cells

Superior
hypophyseal
artery
Stalk

Long portal
vessels
Inferior
Adrenocorticotrophs hypophyseal
artery

Secreting cells

Anterior Posterior
pituitary pituitary

Hormone Short portal


secretion vessel
Glomerulosa
ACTH
Fasciculata

Reticularis

Medulla

Aldosterone Cortisol Androgens Catecholamines

Fig. 1.2 (continued)

derivative (thyroxine), steroid (testosterone), and Peptide hormones are produced by various
lipid (prostaglandin) structures of hormones. endocrine organs or in an ectopic manner by the
Different structures of hormones necessitate cells of certain neoplasms. Peptide hormones act
different receptors. Receptors are then linked to through specific cell membrane receptors; the
intracellular processes that bring about the (in receptors consist of an extracellular domain,
normal situation) desirable effects. Some hor- which directly interacts with the ligand hormone,
mones exert a limited effect on a single organ a transmembrane domain that connects actions
(GnRH) or a few organs, and others have wide- outside the cell with the actions destined to occur
spread effects (thyroxine). Some hormones are within the cell, and the intracellular domain that
stored in their gland of origin (peptide hormones), contains the cellular constituents that cause vari-
while others have limited storage and must be ous biological actions (Fig. 1.3). These internal
synthesized to exert effects (steroids). Many hor- cellular actions include phosphorylation of pep-
mones circulate in a protein-bound state that acts tides or proteins and the generation of other mol-
as a reservoir for future use or limits biological ecules that cause a cascade of events, ultimately
effect (SHBG for sex steroids or growth hor- leading to the metabolic action expected by the
mone- binding protein for growth hormone). presence of the hormone on the receptor (Fig. 1.3).
1 Introduction to Pediatric Endocrinology: The Endocrine System 5

c Third ventricle Hypothalamus


Neuroendocrine
cell nuclei

CRF secreting cells

Superior
hypophyseal
artery
Stalk

Long portal
vessels
Inferior
Adrenocorticotrophs hypophyseal
artery

Secreting cells

Anterior Posterior
pituitary pituitary

Hormone Short portal


secretion vessel
Glomerulosa
ACTH
Fasciculata

Reticularis

Medulla

Aldosterone Cortisol Androgens Catecholamines

Fig. 1.2 (continued)

Table 1.1 The differential diagnosis of primary versus secondary or tertiary endocrine disease or autonomous
function
Target organ hormone
Trophic hormone secretion (e.g., adrenal
secretion (e.g., ACTH) gland) Condition Example
Normal Normal Normal
Low High Autonomous function of Adrenal adenoma
target gland (see Fig. 10.6)
High High Target gland is stimulated by Cushing disease caused by
hypothalamic–pituitary axis pituitary microadenoma
(see Fig. 10.5)
Low Low Secondary or tertiary failure ACTH deficiency (see
of target gland Fig. 1.2c)
High Low Primary failure of target Addison’s disease (see
gland Fig. 1.2b)
6 1 Introduction to Pediatric Endocrinology: The Endocrine System

Cell Insulin IGF-1 GH


membrane
IR IGF1R GHR

JAK2

IRS1 Shc IRS1 Shc IRS Shc

p85 p85
P13K GRB2 p110
P13K GRB2
p110
Akt Akt
STAT5
MAPK MAPK
cascade cascade

NUCLEUS
Protein synthesis IGF-1
glycogen synthesis +
inhibition of SOCS
gluconeogenesis
inhibition of lipolysis Cell proliferation
regulation of and growth
cell survival

Fig. 1.3 Schematic structure of cell-surface heterodi- through binding elements of target genes. SOCS (suppressor
meric insulin/insulin-like growth factor 1 (IGF-1) recep- of cytokine signaling) provides a localized feedback loop
tor and GH receptor (GHR) on an exemplary cell surface. in which the consequences of GH binding to its receptor
The insulin receptor is a heterodimer composed of two α- can be regulated. IGF-1 also provides a localized feed-
and two β-subunits. The type 1 IGF receptor has a similar back loop. The amino acid sequence of the extramem-
structure to the insulin receptor. Both have similar cellular brane portion of the GH receptor is cleaved from the GHR
responses to the ligand binding to the receptor, more the and becomes the circulating GH-binding protein (GHBP)
insulin sequence favors the IRS-1 to Akt pathway while which thereby reflects the abundance of GHRs.
the IGF-1 receptor proceeds through the MAPK pathway. Abbreviations: IGF-1 – insulin-like growth factor 1, GH –
In a situation in which there is excess insulin, such as growth hormone, IR – insulin receptor, IGF-1R – insulin-
found in the infant of a diabetic mother, insulin can cross- like growth factor 1 receptor, GHR – growth hormone
react with the IGF-1 receptor leading to increased growth. receptor, IRS-1 – insulin receptor substrate 1, Shc – Shc
Conversely, IGF-1 can interact with the insulin receptor, protein, GRB2 – growth factor receptor-bound protein 2,
causing insulin effects including hypoglycemia. Two PI3K – phosphatidylinositol 3-kinase, Akt – Akt protein,
independent GH receptor (GHR) molecules must interact JAK2 – Janus kinase 2, STAT5 – signal transducer and
with one molecule of GH to cause intracellular activities. activator of transcription 5, SOCS – suppressor of cyto-
This leads to the recruitment of Janus kinase 2 (JAK2), kine signaling (Modified from Trobec K, von Haehling S,
which phosphorylates the GHR, which provides a dock- Anker SD, Lainscak M. Growth hormone, insulin-like
ing site for STAT (signal transducer and activator of tran- growth factor 1, and insulin signaling-a pharmacological
scription). STAT is then phosphorylated and migrates target in body wasting and cachexia. J Cachexia
uncoupled to the nucleus, where it regulates transcription Sarcopenia Muscle. 2011 Dec;2(4):191–200)

G protein-coupled receptors (including ACTH, have seven transmembrane domains and a G


vasopressin, luteinizing hormone (LH), follicle- protein complex that regulates the second mes-
stimulating hormone (FSH), TSH, gonadotropin- sengers such as calcium and cyclic adenosine
releasing hormone (GnRH), thyroid-releasing monophosphate (AMP). Cytokine receptors for
factor (TRF), growth hormone-releasing factor GH, prolactin, and leptin consist of extracel-
(GHRH), CRF, somatotropin-releasing inhibiting lular, transmembrane, and cytoplasmic domains
factor (SRIF or SS), glucagon, PTH receptors) (Fig. 1.3). The extra membrane domain may
1 Introduction to Pediatric Endocrinology: The Endocrine System 7

Peptide hormone

Peptide ␣s
hormone
receptor ␤ ␥

G-protein ␣s ␣s
active adenyl cyclase
Adenyl cyclase
(inactive) ATP cAMP

A A A A
Phosphorylated CAT CAT CAT CAT
molecules PKA (active) PKA (inactive)

Fig. 1.4 The binding of a peptide hormone to its cell mem- with a specific G protein subunit, in this case, a Gs-alpha
brane receptor, triggering intracellular events which elicit a protein subunit, which converts inactive adenyl cyclase to
response. Note the seven-transmembrane structure of the the active form which in turn catalyzes the conversion of
G-protein-coupled peptide hormone receptor (GPCR) ATP into cyclic adenosine monophosphate (cAMP). cAMP
which in this case is a receptor for glucagon used as an then activates protein kinase A (PKA) by binding to the
example for the general class of membrane-bound recep- regulatory subunits of PKA (noted by an A). The regulatory
tors. When the ligand attaches to its receptor, the GPRC subunits then dissociate from PKA and the remaining cata-
undergoes a conformational change leading to activation of lytic subunits in turn phosphorylate other proteins. Various
the associated heterotrimeric G protein compound by effects can occur in the system based upon the G protein
exchanging its bound GDP for GTP which then associates involved. Adapted from biochemistrypage.org, 2015

reflect the structure of circulating binding pro- receptors may cause disease by rendering the
teins; e.g., the extracellular domain of the growth receptor inoperative, or alternatively, the receptor
hormone receptor (GHR) bears the same amino may be stimulated without the presence of the
acid sequence as a circulating growth hormone- hormone in constitutive activation. The G protein
binding protein (GHBP) with which GH circu- complex may be abnormal in certain diseases;
lates. The cytokine receptors must dimerize (two e.g., McCune–Albright syndrome is associated
receptor molecules must join) to trigger a meta- with constitutive activation of G stimulatory pro-
bolic effect. The intracellular domain causes tein in affected cells. A constitutively active
phosphorylation of tyrosine molecules (JAK2 mutation in the seven-transmembrane domain
kinases), which then phosphorylate signal trans- may be present, as occurs in familial germ cell
ducers, and activators of transcription (signal and Leydig cell maturation. Alternatively, dimer-
transducer and activator of transcription; STAT), ization may fail to occur in some cases of GH
which then travel to the nucleus to regulate deoxy- insensitivity.
ribonucleic acid (DNA) action. The family of insulin receptors includes the
Peptide hormone receptor number and avidity insulin and insulin-like growth factor (IGF)-1
may be regulated by hormones; continuous rather receptors (Fig. 1.4). These are heterodimeric,
than episodic exposure to GnRH downregulates consisting of two α- and two β-chains. When the
GnRH-receptor number as well as receptor activ- appropriate hormone binds to the extracellular
ity on pituitary gonadotropes. This phenomenon domain, the conformation is altered, and phos-
is utilized in treatment with super-active gonado- phorylation of tyrosine occurs in the intracellular
tropin-releasing hormone agonists. Mutations of portion. Through a cascade of effects involving
8 1 Introduction to Pediatric Endocrinology: The Endocrine System

Plasma Steroid hormone


membrane
The unbound steroid hormone
diffuses through the plasma
membrane
Receptor−
Cell
hormone
membrane Receptor
complex The steroid hormone binds to
protein its receptor and enters the
nucleus
Nuclear
membrane Hormone
Nucleus response
elements The steroid hormone−receptor
complex binds to a specific
DNA sequence, the hormone
DNA response element

mRNA Transcription of the gene to


mRNA results from the binding
of the complex to the DNA
Cytoplasm

Specific protein synthesis


New protein results are determined by the
mRNA

Fig. 1.5 Molecular pathway of steroid hormone action. (HREs), or receptor sites, of DNA causing the subsequent
The steroid circulates with a binding protein, from which transcription of DNA into mRNA and translation of the
it dissociates to pass through the cell membrane and reach mRNA into proteins which exert their subsequent effects.
the cytoplasm. The receptor is complexed with heat shock Similar mechanisms are employed by members of the thy-
proteins (not shown), which dissociate, allowing the ste- roid receptor (TR) gene family, though most of the latter
roid to bind to the receptor. Once bound to the steroid, the are concentrated in the nuclear compartment and are not
steroid receptor is activated and is transported into the associated with the heat shock protein (HSP) complex
nucleus. The steroid receptor complex changes conforma- prior to binding with the ligand. Adapted from Studyblue.
tion, allowing it to bind to the hormone response elements com, 2015

insulin-receptor substrates (IRSs), activation of cell wall into the cytoplasm. Specific cytoplasmic
mitogenesis and proliferation as well as effects steroid receptors bind to the steroid molecule, and
on carbohydrate metabolism occur. Insulin and the hormone–receptor complex translocates to
IGF-1 have differing effects on cell growth and the nucleus (Fig. 1.5). The steroid receptor
metabolism, and while each molecule has greater then binds to DNA hormone response elements
effect on its own specific receptor, insulin can to produce the synthesis of messenger RNA
bind to the IGF-1 receptor, and IGF-1 can bind to (mRNA) which leads to translation of the mRNA
the IR exerting effects characteristic of the other on ribosomes and the production of proteins or
molecule. In states of elevated insulin such as in peptides predicted by the steroid hormone that
the infant of a diabetic mother, interaction of the result in changes in cell function.
insulin with a receptor similar to the insulin Thyroid hormone receptors are similar to
receptor, in this case the IGF-1 receptor, occurs steroid receptors in structure and function and are
stimulating growth. members of the steroid hormone superfamily.
Steroid hormones circulate noncovalently The receptors are present either as monomers
bound to various binding proteins. Steroid hor- (TR), heterodimers with retinoid X receptor
mones exert their effects by diffusion through the (TR/RXR), or homodimers (TR/TR). TR/RXR
Suggested Readings 9

heterodimers are the most transcriptionally active Many of the conditions in this book are caused
complex. In the absence of hormones, TR exists by gene mutations. If there is a genetic link for
in a complex with corepressor proteins binding to a given condition, a reference to the Online
hormone response elements (HREs) in DNA in Mendelian Inheritance in Man (OMIM) is placed
an inactive state. Binding of thyroid hormone after the first mention of the condition. Further
results in a conformational change in TR which information can be found on the OMIM website,
displaces corepressors from the receptor/DNA http://www.ncbi.nlm.nih.gov/Omim/. (OMIM
complex while adding coactivator proteins to the can also be found under a pulldown menu tab
complex. Once the DNA/TR/thyroid hormone/ entry on the PubMed site).
coactivator complex is formed, RNA polymerase The format is as follows as described verbatim
transcribes DNA into mRNA which leads to on the OMIM website:
translation of the mRNA on ribosomes and the Each OMIM entry is given a unique six-digit
production of proteins or peptides predicted by number as summarized below:
the hormone that result in changes in cell
function. 1----- (100000-) 2----- (200000-) Autosomal loci
Because of the feedback loops, interpretation or phenotypes (entries created before May 15,
of serum hormone levels must be related to their 1994)
controlling factors; a given value of PTH may 3----- (300000-) X-linked loci or phenotypes
be normal in a eucalcemic patient, but the same 4----- (400000-) Y-linked loci or phenotypes
value may be inadequate in a hypocalcemic 5----- (500000-) Mitochondrial loci or phenotypes
patient with partial hypoparathyroidism, and this 6----- (600000-) Autosomal loci or phenotypes
same value of parathyroid hormone may be (entries created after May 15, 1994)
excessive in a hypercalcemic patient who might
have hyperparathyroidism. Thus a single endo- Allelic variants are designated by the MIM
crine test may be inadequate to evaluate anoma- number of the entry, followed by a decimal point
lies of a feedback loop. and a unique 4-digit variant number. For exam-
A diurnal rhythm of hormone secretion occurs ple, allelic variants in the factor IX gene (300746)
in some systems (e.g., serum ACTH increases are numbered 300746.0001 through 300746.
in the early morning hours, followed by an increase 0101.
in serum cortisol and by a decrease in both during The symbols preceding a MIM number
the afternoon and evening). If the rhythm is dis- represent:
turbed, the amount of hormone present will vary An asterisk (*) before an entry number indi-
from the normal pattern, and disease might occur; cates a gene.
if the normal decrease of cortisol does not occur in A number symbol (#) before an entry number
the evening, Cushing syndrome might result, sim- indicates that it is a descriptive entry, usually of a
ply because the p.m. cortisol values match the a.m. phenotype, and does not represent a unique locus.
values and total daily cortisol secretion is exces- The reason for the use of the number symbol
sive. Thus, although there is little increase in serum is given in the first paragraph of the entry.
cortisol values above normal a.m. values, a great Discussion of any gene(s) related to the pheno-
increase in cortisol effect occurs in such patients type resides in another entry(ies) as described in
affected by Cushing disease. the first paragraph.
Knowing the basic functions of hormones and A plus sign (+) before an entry number
their interactions lends logic to the evaluation of indicates that the entry contains the description
patients with endocrine diseases. This volume of a gene of known sequence and a phenotype.
attempts to emphasize such a systematic evalua- A percent sign (%) before an entry number indi-
tion of endocrine disease. The chapters are based cates that the entry describes a confirmed Mendelian
on organ systems and begin with a brief explana- phenotype or phenotypic locus for which the
tion of the basic physiology at work. underlying molecular basis is not known.
10 1 Introduction to Pediatric Endocrinology: The Endocrine System

No symbol before an entry number generally website which addresses various genetic syn-
indicates a description of a phenotype for which the dromes, some of which are listed in this book.
Mendelian basis, although suspected, has not been The GeneTests website lists genetic clinics and
clearly established or that the separateness of this locations, and genetic tests can be obtained from
phenotype from that in another entry is unclear. research labs or commercial laboratories.
A caret (^) before an entry number means the
entry no longer exists because it was removed
from the database or moved to another entry as Suggested Readings
indicated.
*, The gene location is reliably matched with Online Mendelian Inheritance in Man, OMIM®.
the clinical situation. McKusick-Nathans Institute of Genetic Medicine,
Johns Hopkins University (Baltimore, MD). World
#, Two or more genes can cause the phenotype. Wide Web URL: http://omim.org/ https://www.genet-
Lack of either symbol, no mode of inheritance ests.org/
has been proven. Pagon RA, Adam MP, Ardinger HH, et al.,
The website often has a link to photographs editors. GeneReviews® [Internet]. Seattle (WA):
University of Washington, Seattle; 1993–2015.
demonstrating any dysmorphic characteristics of Available from: http://www.ncbi.nlm.nih.gov/books/
a condition. This source is frequently updated NBK1116/
with clinical and basic information on the sub- Kublaoui B, Levine MA. Chapter 3—Receptor transduc-
jects referenced. There is a useful clinical synop- tion pathways mediating hormone action. In: Pediatric
Endocrinology (Fourth Edition). Sperling MA editor.
sis of many conditions that are listed. The reader 2014: 34–89.
may benefit from frequently accessing this Menon RK, et al. Chapter 2—Molecular endocrinology
resource or the associated GeneTests website. It and endocrine genetics. In: Pediatric Endocrinology
will be apparent if an entry has that many of the (Fourth Edition). Sperling MA, editor, 2014:9–33.
Habener JF. Mechanism of action of hormones that act on
entries have been recently updated with new ref- nuclear receptors. In: Williams textbook of endocri-
erences and new information as indicated by a nology. Kronenberg HM, Mel. Med S, Larsen PR,
date noted at the bottom of the entry; other entries Polonsky KS, editors. Elsevier/Saunders 2011.
may still await such updates and may date from Spiegel AM, Carter-Su C, Taylor SI, Kulkarni
RN. Mechanism of action of hormones that act at the
several years earlier. cell surface. In: Williams textbook of endocrinology.
It is important to remember that many of the Kronenberg HM, Mel. Med S, Larsen PR, Polonsky
autosomal-dominant conditions in OMIM may KS, editors, Elsevier/Saunders. 2011.
actually arise de novo rather than in an inherited Gardner DG, Anderson M, Nissenson RA. Hormones and
Hormone Action. In: Greenspan’s Basic & Clinical
pattern. Endocrinology (9th edition). Gardner DG, Shoback
Two other websites are listed below for genetic DM, Greenspac FS, editors. New York: McGraw-Hill
information. GeneReviews is a peer-reviewed Medical, 2011.
The Evaluation of a Child
or Adolescent with Possible 2
Endocrine Disease

As in all disciplines, the history and physical Medical history must aim to uncover any
examination are crucial in determining a course possible chronic disorders that might have con-
of evaluation. A standard pediatric history and tributed to the disease under evaluation or might
physical examination will serve the evaluator complicate the treatment. Many chronic disor-
well, but in a few areas, increased attention is ders may decrease growth rate; it might be said
required. The type of problem under consider- that almost every page of a pediatric textbook has
ation may change the direction of questioning a reason for impaired growth. If no previous
and evaluation. The following general approach height measurements are available, a history of
is discussed in more detail in the following chap- changes in shoe or clothing size can be of value
ters regarding specific disorders, but this outline to determine whether the child is growing ade-
is meant to direct the initial evaluation. quately, although follow-up measurements over a
period of at least 3 months (and longer if possi-
ble, as accuracy increases with the length of time
The Medical History of follow-up) are necessary for diagnosis. In older
girls, or younger ones if the issue requires it, a
In many cases, the diagnosis is apparent from the menstrual history is necessary; age of menarche
medical history (Table 2.1). In general, the parents (onset), regularity, and amount of flow and dis-
will be the sources of information, but in most comfort are important. Inquire about abnormal
cases, it will be useful to obtain previous medical patterns of urination or defecation.
records, often from several sources because of the The interview to obtain a medication history
recent tendency of transferring care of a child if must often be specific, as many do not consider
the child moves to new locations or has a manda- vitamins or “natural substances” as medicine,
tory change of insurance. Modern electronic although excessive vitamins might be the very
medical records have menus for medical, surgi- cause of a disorder (e.g., hypervitaminosis D). In
cal, family, and birth histories, which should be addition, medications found around the house may
carefully filled out once and easily accessed in be of importance; did the child get into the oral
future visits for reference or updating. contraceptives or hormone replacement therapy
All aspects of the pregnancy history may be of used by another member of the family? Lotions,
importance. This includes medical complica- creams, and hair treatments may contain hor-
tions, nutritional status, toxic or medication mones; tea tree oil may cause feminizing effects
exposures (smoking, infections, medications), while testosterone gel can be transferred from the
gestational age, complications or difficulties of intended recipient to children by touch or by the
delivery, and Apgar scores. use of a common towel. Diet and nutritional

© Springer International Publishing Switzerland 2016 11


D.M. Styne, Pediatric Endocrinology, DOI 10.1007/978-3-319-18371-8_2
12 2 The Evaluation of a Child or Adolescent with Possible Endocrine Disease

Table 2.1 Medical history for pediatric endocrinology


Chief complaint
History of present illness
Birth history and prenatal history
Gestational age
Complications of pregnancy
Toxic exposures
Maternal accidents
Medication taken including vitamins and “natural” treatments
Substances used, including cigarettes
Were ultrasounds obtained, and was the growth normal?
Was fetal motion normal?
Newborn period
Method of delivery
Complications of delivery
Orientation of delivery
Apgar scores
Birth weight % related to gestational age
Developmental assessment
Use of oxygen or other types of support or treatment
Hypoglycemia; documented by blood sugar value or inferred by activity or behavior?
Development
Age of milestones: sitting, cruising, walking; speaking words, sentences. Perform a developmental test if problems
are noted
Family history
Direct questions are often necessary, as family might not volunteer information to general questions. Refer to
chapters for associated conditions of importance
Note full siblings, half siblings, stepparents, or biologic parent relationships
Construct a family tree if possible
Were there any miscarriages?
Age and percentile of height and weight of siblings
Height and weight of parents; approximate weight, if obesity is noted, is important
Ethnicity of parents
Area where parents spent their childhood, especially if there is a possibility of famine, war, or refugee status in
their history
Is there consanguinity?
Age of menarche in mother and sisters if old enough
Age father stopped growing or started to shave and the same information from brothers, if old enough
History of disease similar to patient or otherwise of importance in related individuals
Ask about early deaths due to heart disease or strokes specifically in all conditions under evaluation
Social: who lives at home, what is their relationship, how do they interact, and are there adequate funds for the
child’s benefit especially for nutrition?
Diet: is there adequate food and of a healthful quality? Is there any aversion to eating, is there lack of satiety, or is
there an unusual diet? In evaluation of obesity, much more detail of diet composition and a dietician consultation are
important
Surgical procedures
Allergies
Accidents, especially to the head
(continued)
Physical Examination 13

Table 2.1 (continued)


Medications taken, including vitamins or “natural substances.” Direct questions are often necessary, as family might
not volunteer information to general questions such as, “Does the child take any medications?” Does anyone have
high blood pressure may be answered negatively until you find that the individual is taking medications to make the
blood pressure normal!
School history: grade level, grades obtained, any changes, interrelationships with schoolmates, teasing or bullying
interactions?
Educational achievement in patient and in siblings and parents
Review of systems in general and directed specifically to the issues under consideration
Areas of concern will vary with the disease under consideration, so check the appropriate chapters to determine
specific symptoms and signs of importance
In most situations today, ask about amount of television viewing or screen time per day, amount of activity, sports or
other forms of exercise, and especially do so in children with obesity

patterns are of general importance but also may determine whether consanguinity is present. Are
contribute to the etiology of a condition. Significant there relatives who died young without diagno-
levels of estrogen or other substances might be ses? Indeed in this era of obesity, all cardiac events
found in a noncommercial source of beef. or strokes at a young age (various definitions are
Educational achievement, ability, and history used for “young,” but one suggested is the occur-
are important. In addition, it is important to deter- rence of an event before age 50 years in men and
mine if of psychological problems if related to before age 60 years in women) must be noted.
the condition the child is manifesting (e.g., severe Even though it may be an endocrine visit, queries
short stature or precocious puberty)? about dyslipidemia or hypertension are important
Family history is of great importance in the in this age of obesity. The examiner must ask the
evaluation. Family history of chronic disease, age of menarche in the mother or of the growth
including neurologic or endocrine conditions, is spurt or age at first shaving in the father (a father
determined, and the construction of a family tree will rarely recall his age of onset of puberty!).
is helpful. In many cases, the questions must be Interfamilial interactions can be observed dur-
direct, such as, “Has anyone in the family had thy- ing the interview process to evaluate the possibil-
roid disease?” rather than a general query since ity of psychosocial dwarfism or other psychosocial
many feel that once treated, there is no disease; complications.
asking if someone in the family has hypertension A history of surgery, allergies, and accidents
may elicit “no” as an answer; asking whether some to the central nervous system (CNS) or other
of the family members take medication for hyper- important areas is pertinent.
tension might be returned with the statement
“I do but it is all under control now.” Because par-
ents of pediatric patients are young and subject to Physical Examination
developing new medical conditions (such as type
2 diabetes, hypertension, or dyslipidemia) which Physical examination must be complete (Table 2.2).
are of importance in the evaluation of the child, Accurate determination and plotting of height
family medical history must be obtained repeat- (in centimeters) as described in Chap. 5, weight
edly at subsequent visits. Did the parents immi- (in kilograms), and body mass index (BMI) must
grate from a developing country or live an accompany the determination of vital signs (see
underprivileged life to account for their own his- Chap. 13 for details). An infant should have weight
tory or stature, if abnormal? Was the child and height interpreted in terms of gestational age
adopted, or should another biologic parent who is (by using intrauterine growth charts; see Chap. 5).
no longer in the house be included in the history? A patient who has stature below the third per-
If an autosomal recessive defect is suspected, centile for height according to the Centers for
14 2 The Evaluation of a Child or Adolescent with Possible Endocrine Disease

Table 2.2 Physical examination for pediatric endocrinology


All vital signs will vary with the age of the child, so refer to the Harriet Lane Manual or other sources for standards for
age
Pulse
Blood pressure (interpret in terms of blood pressure for height, and make sure there is an adequate-sized cuff; see
Chap. 13)
Respiratory rate
Temperature if pertinent to disease under consideration or intercurrent illness
Infant length, measured as described in Chap. 5, performed by two adults
After age 2 years, measure height in centimeters on a stadiometer without shoes on; repeat 2 or 3 times if stature is
the main complaint, and make sure measurements are consistent and repeatable within 0.3 cm
Weight in kilograms in light or no clothes
Calculated body mass index (BMI) (kg/m2) interpreted on chart for sex and age
Upper/Lower-segment ratio as necessary (especially in boys with disorders of puberty) (see Chap. 5)
Arm span as necessary (especially in chondrodystrophies or abnormalities of puberty) (see Chap. 5)
HEENT (head, eyes, ears, nose, throat)
Look for midline defects, including cleft palate or lip
Observe for signs of syndromes
Cataracts or colobomas?
Development and status of dentition for age. Single central maxillary incisor as a midline defect that might relate to
hypopituitarism?
Acne or comedones?
Beard?
Voice change?
Neck
Motion
Goiter? Measure width and height of thyroid lobes, and estimate thickness (e.g., 25 % or 50 % greater than normal)
Nodules? Bruits?
Acanthosis nigricans at back of neck?
Lungs: customary pediatric examination
Heart: customary pediatric examination
Abdomen: customary pediatric examination
Axillary hair or odor; enlarging axillary sweat glands?
Breast stage in girls (see Chap. 9)
Stage of pubic hair (see Chap. 9)
Stage of genital development in boys (see Chap. 9)
Extremities: customary pediatric examination unless chondrodystrophy is suspected, and then note ratio of proximal
to distal portions of extremities. Look for Madelung deformity in short stature (see Chaps. 5 and 9). Note motion of
joints and back. Evaluate upper-to-lower segment ratio. Observe for contractures or subtle signs of cerebral palsy.
Scoliosis evaluation
Skin
Café-au-lait spots (number, type, shape, and size), subcutaneous calcifications, acanthosis nigricans
Neurologic: customary pediatric examination with special attention to the CNS in most cases
Cranial nerves: signs of central nervous system (CNS) disease, optic disk development, visual fields, and, in delay of
puberty, sense of smell
Chvostek or Trousseau signs
Tremor
Deep tendon reflexes: customary pediatric examination but look for delayed relaxation of ankle reflexes
Physical Examination 15

Disease Control (CDC) or WHO charts, who is point to a midline defect, syndrome, or neuro-
growing at a rate less than the fifth percentile for logic condition. Is there a goiter or nodule of the
height velocity for age, or is below the third per- thyroid gland? Cardiac, pulmonary, and abdomi-
centile for corrected mid parental height is wor- nal examinations must be thorough but do not
thy of evaluation; a combination of two or more differ appreciably from those in a general pediat-
of these characteristics warrants increased concern. ric examination. Skin examination might reveal
Determination of arm span and upper-to-lower café-au-lait spots or subcutaneous calcifications.
segment ratio is useful in the evaluation of short The extremities may appear to be curved or
stature (e.g., to indicate hypochondroplasia or abnormal or an abnormality of gait may be noted,
achondroplasia or other abnormalities of dispro- as an indication of rickets. Neurologic examina-
portionate growth) or of delay in puberty (e.g., to tion is essential in many disorders considered in
look for the long arms and lower upper-to-lower the book. Signs of dysfunction might suggest a
segment ratio of hypogonadism known as neoplasm or a congenital defect associated with
eunuchoid proportions). The arm span is mea- an endocrine condition.
sured with the patient standing with the back to In almost all conditions, it is important to
the wall with arms spread horizontally and is the determine the stage of pubertal development.
distance from one outstretched middle fingertip This must be done with care and consideration, as
to the other. The lower segment is measured from the patient, especially in the teenage years, may
the top of the symphysis pubis to the floor, be embarrassed; if the patient refuses and the
whereas the upper segment is calculated by sub- caregiver cannot achieve acquiescence, this por-
tracting the lower segment from the height of the tion of the examination may have to be omitted
child. The upper-to-lower segment ratio varies on this visit, as it should not be done by force.
with age (Fig. 5.7). A decreased upper-to-lower Even with cooperation, a nonrelated adult chaper-
segment ratio is found in Klinefelter syndrome, one should be in the room during the examina-
and an increased ratio is found in untreated hypo- tion. Determination of stage of breast and pubic
thyroidism, among other possibilities. hair growth is performed in girls and genitalia
If a problem of growth or pubertal progression and pubic hair (as well as beard) in boys, accord-
is under evaluation, one must consider whether a ing to standard rating techniques (see Chap. 9).
low body weight is the source of the problem. If a child refuses the exam, pictures as in Figs. 9.1,
If nutrition is suboptimal, usually the problem is 9.2, and 9.3 may be presented to the patient to ask
not endocrine in origin, and other causes must be which figure they think they look most like their
considered. Alternatively, if the child is starting stage of development; this is not as accurate as
puberty early or progressing too fast, is excess a detailed exam, but it may be all it is possible.
weight the cause? Every child must have a BMI In addition, the development of axillary odor or
calculated and interpreted on a chart. Body mass hair, the presence of comedones or acne, and the
index charts are found at www.CDC.com; low maturation of facial features are noted in all; if
BMI for age might indicate malnutrition due to the child refuses genital examination, at least this
chronic disease, whereas elevated BMI for age other information is helpful. The appearance of
might indicate rapid growth due to obesity. abnormal distribution or amount of facial or body
The general appearance of the child may fur- hair may be an indication of a problem.
nish a clue as to the chronicity of the problem and Often when no other diagnosis is found in the
its emotional effects. Levels of energy and activ- general clinician’s office, an endocrine disorder
ity are important. Suspicion of a syndrome must is considered and referral made. The results are
be clarified by the examination. The head, eyes, often disappointing as not every obscure symp-
ears, nose, and throat (HEENT) examination may tom or finding can be related to endocrinology!
Disorders of the Hypothalamic–
Pituitary Axis 3

The hypothalamic–pituitary axis might be pic, or hypothalamic–pituitary, portal vessels to


considered as a translator of the action of higher regulate anterior pituitary hormone secretion
central nervous system activity into endocrine (Fig. 3.1). Thus, disorders of any of these intercon-
secretion. In many cases, hypopituitarism is the nected regions, the CNS, the hypothalamus, or the
term used to describe a defect in the secretion of pituitary gland, can cause endocrine disease.
pituitary hormones, but in fact, the condition may A sequence of homeodomain proteins and
be a hypothalamic problem; growth hormone defi- transcription factors cause primordial cells to
ciency most often is caused by the lack of growth develop into the characteristic cells of the ante-
hormone-releasing factor from the hypothalamus rior pituitary gland, the gonadotroph, the thyro-
rather than a defect in the growth hormone- troph, the lactotroph, the somatotroph, and the
secreting cells of the pituitary gland. Thus, hypopi- corticotroph. Mutations in several of these, PROP
tuitarism is a vague term although it is in frequent 1, PIT 1, LHX3, LHX4, HESX1, and TPIT lead
use. to hypopituitarism with a varying array of pituitary
hormone defects depending on the mutation.
There are commercial tests available to screen for
Physiology these mutations.
The pituitary gland receives arterial blood from
The hypothalamus is located in the forebrain the superior hypophyseal artery which gives rise
below the thalamus, hence the name. Under the to the capillary bed that bathes the endocrine cells
influence of higher central nervous system centers of the median eminence of the hypothalamus and
the hypothalamus exerts endocrine effects either into which they release their trophic hormones.
directly, in the production and release of vasopres- These capillaries form the long hypophysiotropic,
sin and oxytocin from the posterior pituitary, or or hypothalamic–pituitary, portal system which
indirectly, through the release of hypothalamic- carries the hypothalamic trophic factors to ensure
releasing or inhibiting factors, small peptides pro- that they reach the cells of the anterior pituitary
duced in minute quantities, which reach the gland to stimulate the secretion of their pituitary
anterior pituitary gland from the median eminence hormones. There is a second short portal system
of the hypothalamus through the hypophysiotro- which connects the posterior and anterior pituitary
glands as well. A second capillary bed drains the
pituitary secretions into the venous system.
The hypothalamic factors are peptides—the
(A more detailed discussion of specific hormones is found
in the chapters related to their functions (e.g., growth hor- shortest, TRF, composed of three amino acids—
mone (GH) is presented in Chap. 5)) that regulate anterior pituitary function. These

© Springer International Publishing Switzerland 2016 17


D.M. Styne, Pediatric Endocrinology, DOI 10.1007/978-3-319-18371-8_3
18 3 Disorders of the Hypothalamic–Pituitary Axis

GRF SRIF or SS GnRH TRF CRF PIF

Hypothalmic
hormone

Effect + – + + + –

GH LH⁄FSH TSH and Prl ACTH Prolactin

Pituitary hormone

Target organ Many tissues, Gonads: Thyroid gland: Adrenal gland: Mammary gland
including the liver, testosterone T4 and T3 Cortisol in females
produce IGF-1 (and DHEAS)

and mammary
gland in females
acting through
or estradiol prolactin

Fig. 3.1 The hypothalamic factors, the pituitary hormones that they control, and the target organs of the pituitary hormones

peptides are growth hormone-releasing factor Prolactin is released in a pulsatile manner and
(GRF) which stimulates the release of growth increases during stress. Dopamine is secreted by
hormone (GH), gonadotropin-releasing hormone tuberoinfundibular dopamine (TIDA) neurons of
(GnRH) which stimulates the release of the the arcuate nucleus and exerts inhibitory effect on
gonadotropins (luteinizing hormone (LH) and the D2 receptors of lactotrophs, causing suppres-
follicle-stimulating hormone (FSH)), thyrotropin- sion of prolactin secretion, and also on the D2
releasing factor (TRF) which stimulates the receptors of the thyrotroph, causing suppression
release of thyrotropin (TSH), and prolactin, corti- of TSH secretion. Estrogen exerts a stimulatory
cotropin-releasing factor (CRF) which stimulates effect on prolactin secretion, and pregnancy is a
the release of adrenocorticotropin (ACTH) and period of increased prolactin secretion. Prolactin
vasopressin, and prolactin inhibitory factor (PIF is high in the neonatal period and then reaches a
or dopamine) which suppresses the release of pro- plateau in childhood only to decrease during male
lactin. Somatostatin (SRIF) has many functions puberty. Prolactin remains stable during female
throughout the body including the gastrointestinal puberty. Like growth hormone, prolactin exerts its
(GI) tract, but for the purposes of this discussion, effects by binding to a cytokine-like receptor on
somatostatin suppresses growth hormone secre- the cell membrane. Prolactin’s most notable endo-
tion as well as TSH and prolactin secretion. The crine effect relates to the production of breast milk
glycoprotein hormones LH, FSH, TSH, and hCG but it exerts many other effects on the immune sys-
are heterodimers composed of an alpha and a beta tem, cell cycle, growth differentiating and anti-apop-
subunit. The alpha subunit is common for all of tosis, hematopoiesis and angiogenesis, and blood
these hormones, but the beta subunits confers clotting.
specificity on each hormone. After secretion into the peripheral circulation,
Prolactin and growth hormone share some the pituitary hormones exert their effects on target
structural homology to human placenta lactogen glands and organs specific for that pituitary
and derive from a common gene of origin. hormone. Target endocrine glands in most cases
Central Nervous System Tumors 19

produce their own hormones that provide feed- Pathology


back to suppress their controlling hypothalamic
and pituitary hormones in turn (e.g., cortisol from Disorders of the hypothalamus or pituitary gland
the adrenal gland provides negative feedback affect endocrine function, but disorders else-
inhibition to the hypothalamic–pituitary system to where in the CNS or radiation therapy to the CNS
suppress hypothalamic corticotropin-releasing for other conditions may also cause significant
hormone that stimulates pituitary adrenocortico- endocrine effects. Although destructive lesions
tropin that in turn stimulates the release of adrenal of the hypothalamic–pituitary axis usually
cortisol). Prolactin is the only pituitary hormone decrease endocrine activity (e.g., hypogonado-
that is mainly suppressed by a hypothalamic fac- tropic hypogonadism), depending on location,
tor, prolactin inhibitory factor (dopamine) and some diseases of the hypothalamic–pituitary axis
GABA (although centrally administered GABA may instead cause increased function (e.g., pre-
may also stimulate prolactin secretion), whereas cocious puberty). Alternatively, functioning
all other pituitary hormones are mainly stimulated lesions may cause endocrine effects because of
by hypothalamic factors (it is true that GH is sup- their secretions rather than exerting effects due to
pressed by the somatotropin release-inhibiting destruction of tissue by their size or location
factor (SRIF), but it is also stimulated by the (e.g., pinealomas secrete the human chorionic
growth hormone-releasing factor (GRF)). Thus gonadotropin (hCG) and cause precocious
hypothalamic disease may lead to a decrease in puberty in boys, while nonfunctioning adenomas
secretion of most pituitary hormones and an of the pituitary gland destroy cell structure and
increase in prolactin secretion, whereas a pituitary can cause decreased pituitary secretion).
gland disorder may cause a decrease in prolactin Specific terminology relates to the level of the
secretion as well as a decrease in other pituitary endocrine lesion. A disorder of the target gland
hormones, a useful diagnostic feature. (e.g., thyroid, adrenal gland) is considered a pri-
The hypothalamus contains the terminal of the mary disease. A lesion of the pituitary gland is
axons of vasopressin-secreting neurons while also considered a secondary defect, and a lesion of the
serving as a location through which other vaso- hypothalamus is a tertiary condition (see Chap. 1,
pressin-secreting axons pass on their way to their Fig. 1.2).
own axon terminal in the posterior pituitary gland.
Thus hypothalamic damage will cause diabetes
insipidus, whereas the result of pituitary stalk sec- Central Nervous System Tumors
tion is variable, depending on the level of lesion. If
a pituitary stalk section or disorder is high on the Most CNS tumors affecting the hypothalamic–
pituitary stalk, all vasopressin-secreting neurons pituitary axis will cause multiple pituitary
may be affected, and the result is diabetes insipi- defects. A hypothalamic tumor may, for example,
dus, whereas if the pituitary stalk section is low, be detected by the presence of galactorrhea, due
some vasopressin-secreting neurons may survive to increased prolactin secretion from the pituitary
intact, and vasopressin secretion and action are still gland in the absence of prolactin inhibitory factor
possible, so that diabetes insipidus may not develop from the hypothalamus, occurring along
or may only be transient (Chap. 4, Fig. 4.4). with deficiencies of other pituitary hormones.
Oxytocin is produced in the hypothalamus and However, in hypothalamic disease, GH is most
transported by neurophysin 1 to and stored in the often affected, and GH deficiency is the most
posterior pituitary. Oxytocin is released with dila- common outcome. GH deficiency may at first
tion of the uterus and cervix and plays a role in nor- appear to be an isolated finding until more careful
mal parturition; exogenous oxytocin is invoked to endocrine evaluation reveals other pituitary
promote the delivery of the baby in certain circum- defects.
stances. Oxytocin plays a role in maternal bonding Because tumors of this area are manifest
with a newborn baby and plays a role in lactation well after birth, compared to congenital defects
after stimulation of the nipples by suckling. which exert effects soon after birth, late onset of
20 3 Disorders of the Hypothalamic–Pituitary Axis

hypothalamic–pituitary deficiencies without Craniopharyngioma appears to be a monoclonal


contributing history (e.g., surgery or trauma to tumor, and about 50 % have genetic abnormali-
the area) may very well indicate the development ties such as gains in activity at 1q, 12q, and 17q.
of a CNS tumor, especially if anterior and poste- About 70 % of cases of craniopharyngioma in
rior deficiencies occur together. In contrast, childhood are the adamantinomatous type with
congenital defects of hypothalamic–pituitary cyst formation. These types have dysregulation
hormones appear at or soon after birth, so that of the Wnt signaling pathway and a mutation in
early onset of combined posterior and anterior the β-catenin gene (CTNNB1) in contrast to the
deficiencies may cause significant effects but do papillary type of craniopharyngioma which has
not necessarily reflect the development of a BRAF mutations and is more often found in adult
tumor. Nonetheless, a magnetic resonance imag- patients.
ing (MRI) evaluation of a child with onset of CNS signs of craniopharyngiomas develop as
hypopituitarism of any age is indicated to deter- the tumor encroaches on surrounding structures.
mine whether a definable anatomic defect is Symptoms of craniopharyngioma include head-
causing the condition. ache, visual disturbances, short stature, diabetes
insipidus, vomiting, and weakness of one or more
limbs. Visual defects including bilateral temporal
Craniopharyngioma field deficits due to impingement on the optic chi-
asm), optic atrophy or papilledema, and signs of
Craniopharyngioma is a rare embryonic malfor- GH deficiency, delayed puberty, and hypothy-
mation of nonglial origin in childhood (0.5–2.0 roidism are features of craniopharyngiomas.
new cases/million population/year or 1.2–4 % Although most patients are shorter than the mean
of pediatric intracranial tumors) but is a more in height and have decreased height velocity at
common CNS neoplasm in pediatrics. It is the diagnosis, a long, indolent course is possible.
most common brain tumor associated with Deficiencies of gonadotropins, GH, thyrotropin
hypothalamic–pituitary dysfunction and sexual (TSH), ACTH, and arginine vasopressin are com-
infantilism and comprises 80–90 % of neo- mon. The serum concentration of prolactin may be
plasms found in the pituitary and up to 15 % of decreased. The bone age reading may be delayed
all intracranial tumors in childhood. Symptoms and is common and may point to the age of onset
usually arise before the age of 20 years with a of tumor growth.
peak incidence between the ages of 6 and 14 About 70 % of patients with a craniopharyn-
years with about 30–50 % occurring in the gioma have suprasellar or intrasellar calcification
pediatric age range. Harvey Cushing introduced (found in fewer than 1 % of normal individuals)
the term “craniopharyngioma” and said that and an abnormal sella turcica, which are some-
they were “the most formidable of intracranial times found on radiographs taken for other indi-
tumors.” cations, including orthodontia. CT reveals fine
Various theories of the embryologic origin of calcifications that are not apparent on lateral skull
this nonglial intracranial tumor are current: one radiographs. MRI scan before and after gadolin-
theory favors development from ectodermal rem- ium is the diagnostic procedure of choice for sus-
nants of Rathke’s pouch and another develop- pected craniopharyngioma and can determine
ment from residual embryonic epithelium of the whether the tumor is cystic or solid and indicate
anterior pituitary gland and of the anterior infun- the presence of hydrocephalus; if necessary, a CT
dibulum. Craniopharyngiomas may reside within scan can be used to search for calcifications. The
or above the sella turcica, or, more rarely, they new susceptibility weighted imaging (SWI) tech-
may be found in the nasopharynx or the third nique allows MRIs to reveal calcifications where
ventricle. older techniques did not.
Central Nervous System Tumors 21

Smaller craniopharyngiomas, usually intrasellar, studies demonstrate the efficacy of bariatric


can be treated by transsphenoidal microsurgery, surgery in the management of obesity in affected
but larger or suprasellar masses which are more patients. Hypothalamic-sparing surgery decreases
frequently found in childhood require craniot- the risk of postoperative hyperphagia and obe-
omy, and the approach must be individualized. sity. Aberrant sleep patterns and even narcolepsy
Shunting may be required for hydrocephalus and daytime somnolence may follow surgical
prior to surgical treatment of the tumor. The treatment of craniopharyngiomas, with melato-
reported post surgical 5-year overall survival is nin improving sleep patterns in some. Although
88–94 %, the 10-year overall survival is 70–92 %, the endocrine complications are more manage-
and the 20-year survival is 76 %. The combina- able, the combination of antidiuretic hormone
tion of limited tumor removal and radiation ther- insufficiency (i.e., diabetes insipidus) and
apy leads to a satisfactory neurologic prognosis, impaired sense of thirst that arises after surgery
better cognitive outcome, and better endocrine in some patients remains a complex management
outcome compared with attempts at complete problem.
surgical extirpation. Frequent and early tumor A Rathke-cleft cyst is often discovered as an
relapse after apparently complete resection and incidental finding on MRI, but it can produce
tumor progression after incomplete resection symptoms and signs indistinguishable from those
suggest the wisdom of radiation therapy after sur- of a craniopharyngioma, such as precocious or
gery. Alternative approaches include proton beam delayed puberty. Surgical drainage and excision
therapy, and, in mainly cystic craniopharyngioma of the cyst wall are customary approaches.
cases, instillation of radioisotopes or sclerosing
substances such as bleomycin or interferon-alpha
is being investigated. Nonetheless, the preferred Germinomas
manner of treatment to retain the best quality of
life is not yet established, but longitudinal studies Germinomas (i.e., pinealomas, ectopic pinealo-
such as the randomized multinational trial mas, atypical teratomas, or dysgerminomas) and
KRANIOPHARYNGEOM 2007 may answer other germ cell tumors of the CNS are the most
this question. It is recommended that craniopha- common extrasellar tumors that arise in the
ryngioma be considered a chronic disease requir- suprasellar hypothalamic region and in the pineal
ing constant monitoring. region that commonly cause sexual infantilism.
The hypothalamic syndrome is comprised of Germinomas constitute 66 % of all intracranial
obesity; fatigue; behavioral changes; circadian germ cell tumor (GCT) which comprise 3–11 %
rhythm irregularities; abnormal sleep patterns; of pediatric brain tumors. About 84 % are found
dysregulation of body temperature, heart rate, in the pineal and the neurohypophyseal regions.
and blood pressure, as well as abnormalities in Peak incidences occur in the second decade and
thirst and is found in approximately one-third of during infancy. They are found more often in
patients at diagnosis in childhood. After surgery, males. Polydipsia and polyuria are the most com-
these findings may occur in 65–80 % of patients. mon symptoms, followed by visual difficulties
Postoperative hyperphagia and obesity (e.g., and abnormalities of growth and puberty or
BMI >5 SD above normal) can be striking and movement disorders. Diagnosis is often delayed
correlate with the magnitude of hypothalamic for months to years because the findings are
damage on cranial MRI. Injury to the hypo- sometimes attributed to psychiatric disorders.
thalamic ventromedial nuclei (associated with Deficiencies of vasopressin and GH are most
increased parasympathetic activity and hyperin- common, but other anterior pituitary hormone
sulinemia) or to the paraventricular nuclei deficiencies (including gonadotropin deficiency)
may cause these findings, and suppression of the and elevated serum prolactin levels are also
increased insulin secretion may be helpful with frequent. Determination of the concentration of
the use of octreotide. Short-term follow-up hCG in spinal fluid and in serum and assessment
22 3 Disorders of the Hypothalamic–Pituitary Axis

of α-fetoprotein levels provide useful tumor Most functional pituitary adenomas are ACTH
markers in children and adolescents with germ secreting, with prolactinomas, GH secreting, or
cell tumors. Germ cell tumors in boys can cause nonfunctioning adenomas occurring less com-
isosexual GnRH-independent sexual precocity by monly. Most pituitary tumors are monoclonal
secretion of hCG (see Chap. 9). Subependymal lesions caused by mutations of guanine
spread of germ cell tumors along the lining of the nucleotide-binding protein, GNAS; a transcript
third ventricle is common, and seeding may of the GNAS gene, Gs-alpha, encodes the alpha
involve the lower spinal cord and cauda equina. subunit of the stimulatory guanine nucleotide-
MRI with contrast enhancement is useful in the binding protein (G protein). Gs-alpha is expressed
detection of isolated enlargement of the pituitary biallelically in nearly all tissues and plays essen-
stalk, an early finding of germinomas that requires tial roles in a multitude of physiologic processes.
periodic MRI monitoring, especially in patients Adolescent onset of pituitary tumors may be the
with diabetes insipidus. The size of the pituitary first manifestation of multiple endocrine neopla-
gland increases by 100 % between year 1 and sia type I (see Chap. 10). Familial isolated
year 15, but the pineal gland does not normally pituitary adenoma (FIPA) (#102200 PITUITARY
change in size after the first year of life; any later ADENOMA) is diagnosed in a family with a his-
enlargement indicates a mass lesion. Pineal cysts tory of pituitary adenomas including prolactino-
are a rare cause of central precocious puberty. mas. About 20 % of cases of FIPA have a germline
Irradiation is the preferred treatment for pure mutation in aryl hydrocarbon receptor-interacting
germ cell tumors such as germinomas; surgery is protein (AIP).
rarely indicated, except for biopsy to establish a With more sensitive imaging techniques,
tissue diagnosis. However, attempts to decrease the presence of a pituitary incidentaloma, a
the long-term morbidity of radiation therapy leads previously unsuspected pituitary lesion that is
to consideration of chemotherapy. Chemotherapy discovered on an imaging study performed for an
alone is inadequate, but the combination of che- unrelated reason, may be discovered. Evaluation
motherapy and radiation therapy can be success- of secretory activity of such a lesion, consideration
ful, and both treatment methods are recommended of mass effects, and follow-up to monitor a change
for a mixed germ cell tumor. Because testicular in size are important, but some lesions detected
germ cell tumors are occasionally found years will not be related to the endocrine abnormality for
after successful therapy for CNS germ cell which the imaging procedure was ordered.
tumors, long-term surveillance is indicated. The incidence of prolactinoma is low in child-
Hypothalamic and optic gliomas or astrocyto- hood, but one in five presents in the 15–24-year
mas, occurring as part of neurofibromatosis (von age group. A case survey reported that 61 % of
Recklinghausen disease (*162200 NEURO- prolactinomas were macroadenomas (more often
FIBROMATOSIS, TYPE I; NF1, due to muta- in boys; hypopituitarism and growth failure were
tions in the neurofibromin gene)) or arising common) and 39 % were microadenomas (more
independently, can also cause sexual infantilism. often in girls; delayed puberty was common).
Gliomas and meningiomas are the most common Delayed onset of puberty was rare, although pri-
CNS tumors to develop in childhood cancer sur- mary amenorrhea was the presenting symptom in
vivors treated with CNS radiation, often in the more than 50 % of pubertal females. Presenting
young adult or even late teenage years. symptoms included oligomenorrhea and galac-
torrhea in the girls and headache in the boys.
Galactorrhea may be demonstrable only by man-
Pituitary Adenomas ual manipulation of the nipples (blood samples
for prolactin should be obtained before examina-
Only 2–6 % of all surgically treated pituitary tion or many hours later, because manipulation of
tumors occur in childhood and adolescence, the nipples raises prolactin levels). The degree of
with about 1 in 1,000,000 children affected. elevation of prolactin may indicate the size of the
Other Central Nervous System Disorders 23

prolactinoma. Microadenomas may demonstrate Other Central Nervous System


prolactin levels over 100 ng/mL, while values Disorders
over 500 ng/mL are suggestive of a macroprolac-
tinoma. Certain medications will also raise pro- Langerhans Cell Histiocytosis
lactin to levels suggestive of an adenoma when in
fact there is no adenoma; risperidone and meto- Langerhans cell histiocytosis (604856 ICD+
clopramide are two examples although there are LANGERHANS CELL HISTIOCYTOSIS) is a
many more psychiatric medications that can raise clonal proliferative disorder of Langerhans his-
serum prolactin values. tiocytes or their precursors characterized by the
Dopaminergic therapy is often successful infiltration of the skin, viscera, and bone with
in decreasing prolactin values. The dopamine lipid-laden histiocytic cells or foam cells. The
agonist bromergocriptine may decrease serum cause is not clear, but there are features of a neo-
prolactin concentrations and decrease tumor size, plasm and features of a reactive immunologic dis-
which is a useful approach before surgery of large order. Diabetes insipidus, caused by infiltration of
macroprolactinomas is undertaken and when the hypothalamus or the pituitary stalk, is the
resection of the adenoma is incomplete. Pubertal most common endocrine manifestation, with GH
progression and normal menstrual function in deficiency and delayed puberty as possible out-
girls usually follow reduction of serum prolactin comes. The lung, liver, and spleen may be
levels. Caberogline is a D2 receptor agonist that involved with this infiltration, and exophthalmos
can be given 2 times per week (not approved for due to infiltration of the orbit is seen. Other find-
children). Pituitary apoplexy followed caberog- ings include cyst-like areas in flat and long bones
line treatment of a macroprolactinoma in a of the skull, the ribs, the pelvis, and the scapula
16-year-old girl; this complication has been seen and the long bones of the arms and legs and in the
in adults treated with bromocriptine. Tricuspid dorsolumbar spine. The appearance of “Floating
regurgitation may occur as a cumulative effect of teeth” within rarefied bone of the mandible and
treatment. Transsphenoidal resection of microp- absent or loose teeth are found. Mastoid or tem-
rolactinomas in children and adolescents is an poral bone involvement may lead to chronic otitis
effective treatment if medication is ineffective. media. Treatment with glucocorticoids, antineo-
High serum levels of macroprolactin, a complex plastic agents, and radiation therapy is promising
of immunoglobulin G and monomeric prolactin in terms of survival, but more than 50 % of
with little biologic activity in vivo, cross-react in patients have late sequelae or disease progression.
commercial prolactin assays, leading to a finding The natural waxing and waning course of this rare
of pseudohyperprolactinemia; high prolactin val- disease makes evaluation of therapy difficult and
ues should be rechecked with subfractionation highlights the importance of national treatment
after polyethylene glycol precipitation to elucidate protocols. Letterer–Siwe disease (%246400
this potential confusion. When prolactin levels are ICD+LETTERER-SIWE DISEASE) has similar-
truly quite high, the molecule may complex with ities to Langerhans' cell histiocytosis but may
IgG antibodies used in certain assays causing a occur in an autosomal recessive pattern.
“hook effect” leading to artifactually low prolactin
levels when values actually are quite high. In this
situation, a 1–100 dilution of serum before analy- Postinfectious Inflammatory
sis is necessary. As prolactin rises in stress, an Lesions of the Central Nervous
upset child may have a falsely elevated prolactin System, Vascular Abnormalities,
suggesting a disorder when there is none. and Head Trauma
It is recommended that screening for multiple
endocrine neoplasia type I (MENIN gene) and Postinfectious or other inflammatory lesions of
FIPA (AIP gene) occur in anyone with a pituitary the CNS, granulomatous disease of the area and
adenoma under 21 years of age. vascular abnormalities, and head trauma may
24 3 Disorders of the Hypothalamic–Pituitary Axis

cause abnormal hypothalamic–pituitary function. central nervous system radiation for a brain
Tuberculous or sarcoid granulomas of the CNS tumor and may be difficult to detect clinically, as
are associated with delayed puberty. The original the growth rate of the child is greater than that
case of adiposogenital dystrophy, or Fröhlich’s found in patients with GH deficiency alone,
syndrome characterized by decreased growth and because the sex steroid secretion increases
delayed puberty with obesity, is thought to have growth rate during pubertal development (see
been caused by tuberculosis infection rather than Chaps. 5 and 9). Irradiation of the CNS in early
a neoplasm. life predisposes the patient to later onset of sec-
Hydrocephalus may cause hypopituitarism, ondary CNS tumors sometimes in just a few
but precocious puberty also is a possible result, years after treatment of the first tumor. Such
depending on the amount of pressure exerted on patient must be monitored to determine what
various central nervous system locations. Clinical pituitary defects have developed so that it may be
findings will include signs of increased intra- treated appropriately.
cranial pressure. When hydrocephalus or sub-
arachnoid cysts, which can cause similar effects,
are decompressed, pituitary abnormalities may Developmental Defects
improve. of the Midline

Congenital defects of pituitary secretion also may


Irradiation of the Central result from anatomic malformations of the hypo-
Nervous System thalamus, from pituitary hypoplasia or aplasia, or
from more subtle defects of hormone secretion.
Radiation of the head for treatment of CNS Congenital defects of the midline associated with
tumors, leukemia, or neoplasms of the head and hypopituitarism range from holoprosencephaly
the face, in which the radiation field involves the (cyclopia, cebocephaly, orbital hypotelorism) to
hypothalamus or pituitary field, may result in the single maxillary incisor and even to cleft palate
gradual onset of hypothalamic–pituitary failure (4 % of cases of cleft lip and/or palate are associ-
over a period of months to a few years. This ated with GH deficiency, so that all growth failure
occurs mainly due to hypothalamic rather than in cleft palate should not necessarily be attributed
pituitary damage by the radiation and is dose- to poor feeding or nutrition alone). Individuals
dependent. This etiology comprises an growing with myelomeningocele (myelodysplasia) have
group of patients with hypopituitarism because an increased frequency of endocrine abnorma-
of the increasing success in radiation treatment of lities, including hypothalamic hypothyroidism,
such neoplasms. GH deficiency is the most com- hyperprolactinemia, and decreased gonadotropin
mon hormone disorder resulting from radiation concentrations, whereas some patients demon-
of the CNS, but gonadotropin deficiency, hypo- strate central precocious puberty.
thyroidism, and decreased bone density also
occur. Self-reported fertility was reported to be
lower in women who received CNS radiotherapy Septo-Optic and Optic Dysplasia
for acute lymphoblastic leukemia at about the
time of menarche, although the average age of Septo-optic dysplasia (optic nerve hypoplasia,
women in a long-term study was in the early 20s, absent septum pellucidum, or variations of
and longer follow-up of fertility may change the both caused by abnormal development of
results. Conversely, early or central precocious the prosencephalon) (# 182230. SEPTOOPTIC
puberty may occur after radiation therapy for DYSPLASIA due to a mutation at 3p21.2-p21.1).
CNS lesions. The combination of GH deficiency The HESX1 homeobox gene or SOX2 (see
and precocious puberty is a possible outcome of below), SOX3, and OTX2 may be associated with
Idiopathic Hypopituitary Dwarfism 25

significant visual impairment which often leads The Solitary Median Maxillary
to pendular nystagmus (to-and-fro nystagmus Incisor Syndrome
due to inability to focus on a target). Small, pale
optic disks occur, but not the appearance of (#147250 SOLITARY MEDIAN MAXILLARY
optic atrophy, which would suggest previous CENTRAL INCISOR; SMMCI) Solitary median
development of the optic nerves with subsequent maxillary incisor is associated with the eponymous
deterioration due to acquired pathology. midline defect and with a prominent midpalatal
A midline hypothalamic defect may lead to ridge (torus palatinus) and hypopituitarism. The
the combination of GH deficiency and diabetes defect in this autosomal dominant condition is in the
insipidus and may be associated with deficient sonic hedgehog gene (SHH) at gene map locus 7q3.
ACTH, TSH, and gonadotropin secretion as well.
Short stature and delayed puberty may be the
most obvious results, although true precocious Idiopathic Hypopituitary Dwarfism
puberty is an alternative outcome in rare cases of
this midline defect. The septum pellucidum is In addition to HESX1 homeobox mutations, auto-
absent in about 50 % of cases of optic hypoplasia somal recessive mutations in homeobox genes
or dysplasia, and this defect is readily demon- encoding transcription factors involved in the
strable by CNS imaging techniques, most fre- early aspects of pituitary development lead to
quently by MRI. hypogonadotropic hypogonadism and other pitu-
The MRI findings of congenital hypopituita- itary hormone deficiencies (Table 3.1). PROP1
rism may include an ectopic posterior pituitary mutations (#262600 PITUITARY HORMONE
gland “hot spot” and the appearance of what DEFICIENCY, COMBINED, 2; CPHD2) at gene
appears to be a “pituitary stalk transection” and map locus 5q35.3 cause GH and TSH deficiency
a hypoplastic pituitary gland due to the lack and produce delayed puberty or late onset of sec-
of hypothalamic stimulatory factors. In some ondary hypogonadism in adulthood and more
patients, the neurohypophysis may appear rarely may also cause ACTH deficiency. In one
absent because of a missing hot spot although study of 73 patients with idiopathic multiple pitu-
physiologic changes in hydration may affect the itary hormone deficiencies, 35 had a mutation in
hot spot, which represents vasopressin content, PROP1. Homozygous Arg73Cys mutation of
with no true anatomic defect. Abnormalities of PROP1 allowed spontaneous puberty in 2 of 10
the corpus callosum and cerebellum are com- affected family members. ACTH deficiency may
mon on MRI. Four groups are described: those develop later and is more rarely a feature of
with normal MRI results, those with abnormali- PROP1 deficiency; patients must be monitored
ties of the septum pellucidum and with a normal for this serious complication.
hypothalamic–pituitary area, those with abnor- Mutations of POU1F1 (PIT *173110
malities of the hypothalamic–pituitary area and POU DOMAIN, CLASS 1, TRANSCRIPTION
a normal septum pellucidum, and those with FACTOR 1; POU1F1 at 3p11) cause deficiency
abnormalities in both areas. No endocrine in GH, TSH, and prolactin.
abnormalities were described in the first group, Homozygous mutations occur in the LHX3
but the others had progressively more endocrine gene (#221750 PITUITARY HORMONE
abnormalities, with precocious puberty most DEFICIENCY, COMBINED, 3; CPHD3) at gene
common in the second group. Early diagnosis is map locus 9q34.3. The LHX3 gene encodes a
important because of the risk of sudden death member of the LIM class of homeodomain pro-
associated with adrenal insufficiency if ACTH teins, which are associated with multiple pituitary
is inadequate. hormone deficiencies, including LH and FSH,
Delayed puberty is rarely described in dupli- and often with severe restriction of head rotation.
cation of the hypophysis. LH4 (#262700
26

Table 3.1 Genetic forms of multiple pituitary/hypothalamic hormone deficiencies


Defect Hormones deficient Inheritance Mutations Other features
POU1F1 GH, Prl, TSH Auto-recessive and autosomal Missense, nonsense, Severe impairment of postnatal growth, variable
dominant frameshift, splicing pituitary hypoplasia
PROP1 GH, Prl, TSH, LH, FSH, Auto-recessive Nonsense, frameshift, Transient pituitary hyperplasia in some
and ACTH developing later splicing
in some
HEX1 Any or all, including Auto-recessive and autosomal Missense, frameshift Septo-optic dysplasia or hypoplasia
vasopressin dominant
LIM HOMEO BOX GH, Prl, TSH, LH, FSH, Auto-recessive Missense, nonsense, Limited neck rotation, short cervical spine,
GENE 3; LIM3 LHX3 and ACTH in some frameshift, splicing sensorineural deafness
LIM HOMEO BOX GH, Prl, TSH, LH, FSH, Autosomal dominant Missense, frameshift Cerebellar abnormalities, very small sella turcica
GENE 4; LIM4 LHX4 and ACTH in some, abnormal petrous bone
SOX2 LH,FSH, variable GH Autosomal dominant Missense, nonsense, Anophthalmia/microphthalmia, esophageal
frameshift atresia, genital tract abnormalities, hypothalamic
hamartoma, sensorineural hearing loss, diplegia
GLI2 GH, Prl, TSH, LH, FSH, Haploinsufficiency Missense, frameshift Holoprosencephaly, craniofacial abnormalities,
3

and ACTH polydactyly, partial agenesis of the corpus


callosum
FGF8 LH, FSH, and vasopressin Auto-recessive and autosomal Missense, chromosome Anosmia, holoprosencephaly, Moebius
dominant deletion syndrome, septo-optic dysplasia
SOX3 Isolated GHD or GH, Prl, X-linked Variations in polyalanine Mental retardation, infundibular hypoplasia,
TSH, LH, FSH, and ACTH tract length, chromosome ectopic posterior pituitary, midline abnormalities
duplication
OTX2 Isolated GHD or GH, Prl, Autosomal dominant Missense, nonsense, Anophthalmia/microphthalmia, coloboma,
TSH, LH, FSH, and ACTH microdeletion developmental delay
TBX19 ACTH Autosomal recessive Missense, nonsense, Neonatal hypoglycemia, neonatal cholestatic
frameshift, splicing jaundice
GH growth hormone, Prl prolactin, TSH thyroid-stimulating hormone, LH luteinizing hormone, FSH follicle-stimulating hormone, ACTH adrenocorticotropic hormone
Disorders of the Hypothalamic–Pituitary Axis
Suggested Readings 27

PITUITARY HORMONE DEFICIENCY, Suggested Readings


COMBINED, 4; CPHD4) and GLI2 (#610829
HOLOPROSENCEPHALY 9; HPE9) mutations 1. Romero CJ, Pine-Twaddell E, Radovick S. Novel
mutations associated with combined pituitary hormone
may cause isolated GH deficiency or combined deficiency. J Mol Endocrinol. 2011;46(3):R93–R102.
pituitary hormone deficiencies including 2. Tsai SL, Laffan E, Lawrence S. A retrospective
gonadotropin deficiency. The X-linked form review of pituitary MRI findings in children on growth
of hypopituitarism can be associated with hormone therapy. Pediatr Radiol. 2012;42(7):
799–804.
duplication of the SOX3 gene (#312000 3. Deal C, Hasselmann C, Pfaffle RW, Zimmermann
ICD + PANHYPOPITUITARISM, X-LINKED; AG, Quigley CA, Child CJ, Shavrikova EP, Cutler Jr
PHPX). Deficiency of SOX2 (#206900 GB, Blum WF. Associations between pituitary imag-
ICD+ MICROPHTHALMIA, SYNDROMIC 3; ing abnormalities and clinical and biochemical phe-
notypes in children with congenital growth hormone
MCOPS3), a transcription factor involved in early deficiency: data from an international observational
hypothalamic–pituitary embryonic development, study. Horm Res Paediatr. 2013;79(5):283–92.
leads to anterior pituitary hypoplasia. Patients with 4. Cavarzere P, Biban P, Gaudino R, Perlini S, Sartore
SOX2 mutations have major eye abnormalities, L, Chini L, et al. Diagnostic pitfalls in the assessment
of congenital hypopituitarism. J Endocrinol Invest.
including anophthalmia, microphthalmia, and col- 2014;37(12):1201–9.
oboma. They also have hypogonadotropic hypogo- 5. Castinetti F, Reynaud R, Quentien MH, Jullien N,
nadism (HH) as the most common pituitary defect, Marquant E, Rochette C, et al. Combined pituitary
in contrast to most other types of pituitary hypopla- hormone deficiency: current and future status.
J Endocrinol Invest. 2015;38(1):1–12.
sia, demonstrating GH deficiency most frequently. 6. McCabe MJ, Dattani MT. Genetic aspects of hypotha-
There is an association between breech delivery lamic and pituitary gland development. Handb Clin
(especially for male infants), perinatal distress, and Neurol. 2014;124:3–15.
idiopathic hypopituitarism. Malformations of the 7. Keil MF, Stratakis CA. Pituitary tumors in childhood:
update of diagnosis, treatment and molecular genet-
pituitary stalk demonstrable by MRI are common ics. Expert Rev Neurother. 2008;8(4):563–74.
in these patients. Other types of birth trauma or 8. Xekouki P, Azevedo M, Stratakis CA. Anterior pitu-
complications may lead to hypopituitarism as well. itary adenomas: inherited syndromes, novel genes and
Common to many patients with congenital hypo- molecular pathways. Expert Rev Endocrinol Metab.
2010;5(5):697–709.
pituitary dwarfism is early onset of growth failure; 9. Griffith R Harsh, Lawrence D Recht, Karen J Marcus.
late onset of diminished growth is an ominous find- Craniopharyngioma. In: UpToDate. http://www.upto-
ing, suggesting the presence of a CNS tumor. date.com/contents/craniopharyngioma.
The Posterior Pituitary Gland
and Disorders of Vasopressin 4
Metabolism

The posterior pituitary gland, also known as the excessive drinking, to urinary tract defects in con-
neurohypophysis, may share a term in its name centrating ability, possibly due to urinary tract
with the anterior pituitary gland but unlike the infections, as well as to osmotic diuresis. The most
anterior pituitary gland has a direct connection to common etiology of a pathologic cause of polyuria
the brain as the posterior pituitary contains the is the glucosuria of diabetes mellitus (translated as
nerve endings of the vasopressin and oxytocin “sweet urine”) in contrast to the rarer condition of
neurons which originate in the periventricular diuresis due to the lack of ADH that leads to central
and supraoptic nuclei. Vasopressin is secreted diabetes insipidus (DI; “weak urine”). Diabetes
from the posterior pituitary gland as is oxytocin. mellitus is the province of Chap. 11.
Disorders of vasopressin metabolism may cause
increased or decreased urine formation and sub-
sequent abnormalities in body water and serum Normal Vasopressin Physiology
osmolality and electrolytes. Rather than being
controlled by secretions of the median eminence Arginine vasopressin (AVP) is the human antidi-
of the hypothalamus through the hypophysiotro- uretic hormone (ADH) (Fig. 4.1). AVP is pro-
pic portal system, these nerve endings in the pos- duced in the magnocellular cells of the
terior pituitary directly secrete their product into paraventricular and supraoptic nuclei of the hypo-
the general circulation. The axons of these neu- thalamus in a large precursor molecule coded by a
rons may be interrupted by trauma or surgery, and gene at 20p13 along with neurophysin II (NPII).
the level of the interruption determines the out- The two molecules are synthesized within the
come. If the pituitary stalk section is low, some neurosecretory granules of the magnocellular
intact vasopressin neurons will remain as some cells and then reassembled into AVP–NPII com-
terminate at the top of the pituitary stalk and dia- plexes before secretion. There are about 30–50
betes insipidus may be mild or not occur. If the days’ worth of AVP stored for secretion in normal
stalk section is high, it is more likely that all vaso- circumstances, but with maximal use, only a
pressin neurons will be affected and that diabetes 5–10-day supply is available. Neurophysin I is a
insipidus will be permanent. molecule of unknown function which is secreted
Although lack or excess of antidiuretic hormone with and transported with oxytocin.
(ADH) is a main focus of the chapter, nephrogenic The magnocellular neurons terminate, for
diabetes insipidus which presents in a relatively the most part, in the posterior pituitary gland
similar manner is also covered. Further, excessive (the neurohypophysis), but some terminate in
urination also may be due to the desire for second- the third ventricle, and some terminate high in the
ary gain in psychogenic polydipsia, to habitual pituitary stalk or in the median eminence of the

© Springer International Publishing Switzerland 2016 29


D.M. Styne, Pediatric Endocrinology, DOI 10.1007/978-3-319-18371-8_4
30 4  The Posterior Pituitary Gland and Disorders of Vasopressin Metabolism

+
Osmoreceptors and Water
sense of thirst Intake

+

Hypothalamus and + + –
pituitary gland
+ + Decreased
blood pressure/
Heart
– volume

AVP – Increased
Increased
serum osmolality blood pressure/
+ + volume

aldosterone +

Adrenal angiotensin Decreased


gland serum osmolality

renin
+
+ Concentrated
Kidney
urine

Fig. 4.1 Regulation of serum sodium and water. Solid vasopressin. Increased blood pressure or blood volume
lines indicate a positive effect and dashed lines indicate a will cease the stimulation of the volume and baroreceptors
negative effect. Increased serum osmolality stimulates the in the cardiovascular system from simulating vasopressin
osmoreceptor and sense of thirst, causing an individual to release, allowing appropriately increased urinary excre-
take in more water and stimulates the release of arginine tion. Low extracellular volume or hyponatremia will stim-
vasopressin (AVP) from the posterior pituitary gland. AVP ulate the kidney to activate the renin–angiotensin system
stimulates the kidney to concentrate the urine by making to stimulate the adrenal gland to produce aldosterone
the distal collecting tubule permeable to water and thus which will retain sodium and increase serum osmolality.
decreases serum osmolality which thereby decreases water Concentration of sodium in the renal tubule will also tend
intake and decreases sense of thirst. Decrease in blood to decrease urinary concentration and volume. Increased
pressure or blood volume will likewise stimulate the blood pressure or volume causes the heart to release atrial
osmoreceptors and volume or pressure sensor in the car- natriuretic peptide which antagonizes the aldosterone
diovascular system to stimulate the release of arginine affect and allows urinary sodium loss

hypothalamus. Vasopressin also is produced in changes in plasma osmolality as subtle as 1–2 %,


the parvocellular cells of the paraventricular reaching a sensitivity that is better than many
nuclei, as is corticotropin-releasing hormone laboratory tests can accomplish. Serum osmolality
(CRH), and both stimulate the secretion of adre- is determined by osmotically active substances in
nocorticotropic hormone (ACTH); in this situa- the blood which include sodium chloride (unless
tion, the vasopressin neurons do terminate in the glucose is significantly elevated). An increase in
median eminence, and their secretions are carried osmolality due to dehydration, or the infusion of
through the portal system to the pituitary gland. a hypertonic solution such as concentrated saline,
An intracellular osmotic detector (organum vas- triggers the release of sufficient vasopressin to
culosum of the lamina terminalis and anterior cause the kidney to retain water, thereby decreas-
hypothalamus) is located close to the supraoptic ing the serum osmolality to normal. When plasma
nuclei but separate from the nuclei containing osmolality is less than 280 mOsm/kg, vasopres-
ADH; the osmotic detector may reside outside of sin is not released to any appreciable degree, val-
the blood–brain barrier. This sensor detects ues range from 0.5 to 1 pg/mL, but when
Normal Vasopressin Physiology 31

osmolality reaches 283 mOsm/kg or more, vaso- inhibit the ability to excrete water and urine and
pressin is secreted in increasing amounts, until may lead to overhydration. Exogenous medica-
maximum secretion occurs at 320 mOsm/kg tions that may inhibit the ability to excrete adequate
which leads to a serum level of about 20 pg/mL. urine volume include medications which are used
Low-pressure baroreceptors in the right atrium in children and adolescents such as lisinopril
and pulmonary venous circulation and high-­ which is used in diabetic nephropathy, carbam-
pressure baroreceptors in the carotid sinus and azepine and valproic acid, sulfonylureas, tricyclic
aortic arch also regulate the release of vasopres- antidepressants, and selective serotonin uptake
sin, so that a decrease in blood volume of 8–10 % inhibitors among others.
(equivalent to a major hemorrhage) will stimulate Vasopressin exerts two major biologic effects:
a large release in vasopressin. Lung disease and it increases permeability of the collecting duct of
respiratory support with ventilator therapy will the nephron to water filtered in the urine and, in
trigger vasopressin secretion because of the large concentrations, ADH stimulates the contrac-
increase in intrathoracic pressure, as detected by tion of arterial muscle, which increases blood
the stretch receptors of the atrium; increased fluid pressure (BP), hence the name vasopressin. V2
management as is often used in pneumonia, receptors located in the renal collecting duct
example, may trigger the syndrome of inappro- mediate the actions of vasopressin on urine con-
priate secretion of ADH (SIADH; see later). centration. Of the three vasopressin receptors, the
During the daily routine of standing and walking, membrane-bound V2 vasopressin receptor (V2R),
vasopressin concentrations change from moment a G-protein-coupled receptor encoded by the X
to moment because of stimulation of the carotid chromosome on the basolateral membrane of
baroreceptors. In states of dehydration and low renal collecting duct epithelial cells, is the most
plasma volume, vasopressin is released by physi- important in water balance. Increased intracellu-
ologic stimulation. If vasopressin is released at lar cyclic adenosine monophosphate (cAMP)
times of elevated plasma volume, this release is results after V2R activation, which mediates shut-
inappropriate. tling of the water channel aquaporin 2 (AQP-­2) to
Vasopressin may be released in other situa- the apical membrane of collecting duct epithelial
tions regardless of plasma volume. Other stimuli cells, resulting in increased water permeability
of vasopressin secretion include nausea and cer- and antidiuresis. Other receptors allow vasopres-
tain drugs, such as chlorpromazine and antime- sin to exert other effects. The V1 receptor is found
tabolites, used in the treatment of cancer; nausea in the vasculature, in the platelets, and in the liver.
in cancer treatment should therefore be an The V3 receptor is found in the anterior pituitary
expected stimulus for vasopressin secretion and gland where it modulates the effects of vasopres-
may cause the syndrome of inappropriate secre- sin on ACTH secretion.
tion of ADH if fluids are administered in excess The medulla of the kidney normally maintains
(SIADH; see later). Nicotine exerts its effects a high osmotic concentration of urea and sodium
through the precipitation of nausea, which stimu- built up by the countercurrent exchange system.
lates the release of vasopressin. When vasopressin allows water filtered in the
There are several situations in which vaso- urine to pass through the walls of the collecting
pressin secretion is increased and the patient has ducts, the water will, through the osmotic gradi-
an inability to excrete water load. Vasopressin ent, be drawn to the medulla, and the urine in the
secretion is inhibited by glucocorticoids and in lumen of the collecting ducts will become more
Cushing’s syndrome urination increases; alterna- concentrated. The effects of vasopressin on salt
tively, decreased glucocorticoids increase vaso- balance are mostly mediated through its effect on
pressin secretion, and in Addison’s disease the changes in water balance as fluid retention will
individual’s inability to excrete a water load may lower serum sodium although vasopressin can
lead to overhydration with a subsequent decrease activate the epithelial Na(+) channel (ENaC) to
in serum sodium. Like­wise, ­hypothyroidism will decrease sodium excretion to a lesser effect. Thus
32 4  The Posterior Pituitary Gland and Disorders of Vasopressin Metabolism

if water is lost, serum sodium and osmolality heart failure that can be caused by volume excess
increase, whereas if water is retained, serum are well appreciated. However, the effects of rapid
sodium and osmolality decrease. fluid shifts on brain cells and brain function can
If vasopressin from the posterior pituitary is not be devastating and may be permanent, and these
produced or cannot be released, central diabetes may occur because of DI or SIADH. A rapid
insipidus (DI) develops. If the collecting duct increase in plasma osmolality can draw fluid from
cannot respond to the vasopressin, nephrogenic the brain and, especially in young infants, cause
DI occurs. If a patient has been drinking large brain shrinkage and rupture of veins that bridge
amounts of water for a prolonged period because the distance from the rigid cranium to the more
of habit or psychogenic polydipsia, the medullary malleable brain substance. Although the brain is
interstitial gradient becomes progressively more slower to correct osmolar balance than is the vas-
dilute (“washes out”), so that the maximal con- cular compartment, during a more gradual increase
centrating ability of the kidney is decreased, and in intravascular osmolality, the brain can produce
polyuria results. Infection or various types of “idiogenic osmoles” (or osmolytes including tau-
kidney disease also can decrease the concentrat- rine, glycine, glutamine, sorbitol, and inositol) that
ing ability of the kidney (causing functional increase intracellular osmolality to balance an
nephrogenic DI). intravascular hyperosmolality. These osmolar-
The sensor responsible for the sense of thirst active molecules rise within 4 h of increased extra-
is located in the lateral hypothalamus. The sensa- cellular osmolality and remain in the brain for
tion of thirst arises when serum osmolality hours to days after a decrease in intravascular
increases to more than 293 mOsm/kg, a 2–3 % osmolality occurs, leading to an imbalance of
rise from normal, which is 13 mOsm/kg higher osmolar forces with the net shift of fluid toward
than the lower limit of stimulation of vasopressin the brain if serum osmolality decreases, result-
secretion. A decrease of 1–4 % of blood volume ing in cerebral edema that could cause cerebellar
will likewise trigger the sensation of thirst. Other tonsillar herniation and death. Thus both hypoos-
sensors in the oral cavity interact with both the molality and hyperosmolality are damaging to
sense of thirst and vasopressin secretion, so that the brain.
fluid ingestion is modulated quite closely, as The renin–angiotensin system also affects
appropriate fluid balance is essential for life. water balance as it affects sodium and potassium
Thus two basic mechanisms control serum balance described in Chap. 10. Decreased intra-
osmolality: (a) vasopressin secretion exerting vascular volume stimulates the renin–angiotensin
effects on the nephron and (b) fluid dynamics system leading to increasing blood pressure,
mediated by fluid loss and the sense of thirst that increasing potassium excretion, and increasing
affects fluid intake. Either mechanism can com- sodium resorption from the distal tubule whereby
pensate for a defect in the other, but if both mecha- water is likewise retained. Manipulation of the
nisms fail, and the individual has neither sense of renin–angiotensin system is one manner of treat-
thirst nor ability to concentrate urine, swings in ing nephrogenic diabetes insipidus (see below).
osmolality may reach a life-threatening degree. An The family of three natriuretic peptides plays
infant is dependent on others to supply water and a role in total body sodium retention. Atrial natri-
may not be able to communicate its needs, leading uretic peptide (ANP) is produced not only in the
to overhydration or underhydration. An older child atrium but in the brain and other areas of the cen-
who lacks vasopressin can usually drink enough tral nervous system and serves to decrease blood
water to maintain fluid balance for a while if the pressure and limit cardiac hypertrophy. B natri-
child’s thirst mechanism operates normally. uretic peptide (BNP due to initial isolation from
The consequences of hyperosmolality or the brain) is produced in the ventricle of the heart
hypoosmolality can be severe. The obvious as well as the brain and acts in a similar manner
impairment of renal function due to severe vol- to ANP. C natriuretic peptide (CNP) which is
ume constriction and the pulmonary edema and produced in the central nervous system also
­
Diabetes Insipidus 33

r­egulates bone growth. While there are three d­isorders. Most commonly in the pediatric age
receptors for the atrial natriuretic peptides, the group, a craniopharyngioma or germinoma is
atrial natriuretic peptide receptor A is located in involved. The germinoma may be associated with
the kidney, interacts with ANP and BNP, and elevated beta hCG levels (or a positive pregnancy
most directly affects sodium balance. ANP is test due to elevated hCG) leading to peripheral
secreted in response to stretching of the atrial ­precocious puberty. Langerhans cell histiocytosis
wall due to increased intravascular volume and comprises several conditions characterized by infil-
BNP is secreted in response to stretching of the tration of tissues by cells similar to Langerhans cells
ventricular wall in states of congestive heart fail- from the bone marrow as well as various hemato-
ure. When ANP binds to the atrial natriuretic pep- poietic cells, including T cells, macrophages, and
tide receptor A, diuresis ensues with natriuresis eosinophils: Langerhans cell histiocytosis (604856
as a result of antagonistic effects on the renin–­ ICD+ LANGERHANS CELL HISTIOCYTOSIS)
angiotensin system; vasorelaxation and increased and Letterer–Siwe disease (%246400 ICD+
endothelium permeability also occur. LETTERER-SIWE DISEASE) are examples
which are associated with DI. The late onset of pos-
terior pituitary disease, well after birth, manifest by
Diabetes Insipidus DI is an urgent reason for a full effort to diagnose a
tumor or infiltrative lesion; if this is associated with
Central Diabetes Insipidus anterior pituitary deficiency, the concern rises fur-
ther. The presence of a thickened pituitary stalk in a
The inability to release adequate arginine vaso- patient with central diabetes insipidus may be an
pressin in the face of increased serum osmolality early indicator of the developing germinoma; it
can be caused by central nervous system (CNS) may take years of observation before the second
tumor, trauma, infection, or granuloma as well as pituitary stalk grows further to reveal the tumor.
by a congenital defect of the brain structure Trauma, whether accidental, such as a fall off a
(Table 4.1; Fig. 4.2) or mutations in the AVP-­NPII horse or an automobile accident, or iatrogenic pro-
gene all leading to central diabetes insipidus. cedures, such as surgery near the posterior pitu-
Some vasopressin secretion may remain in spite itary for a craniopharyngioma, can lead to DI.
of DI, but the amount of vasopressin secreted is Hydrocephalus or other types of increased intra-
not commensurate with the need for water conser- cranial pressure also can lead to vasopressin defi-
vation, and excessive urination occurs. Depending ciency and DI. Radiation to the central nervous
on the nature of the defect, the sense of thirst may system will cause a deficit in various releasing fac-
or may not remain intact, determined by whether tors, but it does not cause diabetes insipidus.
the thirst center of the CNS is affected as well as Idiopathic, congenital DI without anatomic
the vasopressin neurons. If impairment of the abnormalities can occur sporadically, in an auto-
sense of thirst occurs, maintenance of serum somal dominantly inherited familial pattern
osmolality in the normal range becomes most (#125700 ICD+ DIABETES INSIPIDUS,
difficult, as voluntary water intake must replace NEUROHYPOPHYSEAL at 20p13) with facial
previously automatic functions, sensing thirst and abnormalities such as hypertelorism and low bone
drinking when thirsty. Home management of the density or can occur in an autosomal recessive or
patient with diabetes insipidus and a loss of thirst X-linked pattern (304900 DIABETES
sensation is complicated by the fact that there is INSIPIDUS, NEUROHYPOPHYSEAL TYPE)
no home device to measure serum sodium or because of defects in the gene for AVP-­NPII. A
serum osmolality as there is for glucose with a recent prospective study demonstrated that the
home glucometer. Frequent laboratory visits are majority of cases with “idiopathic central diabetes
necessary in such patients. insipidus” actually had an autoimmune basis to
Any hypothalamic–pituitary tumor can cause DI the condition. The authors found that after the
with or without other hypothalamic–pituitary identification of diabetes insipidus, multiple
34

Table 4.1  Causes of disturbances of osmolality and serum sodium


Plasma
anti-­
Urine Serum Serum natriuretic
Condition Serum Na Urine Na Plasma Osm Osm AVP PRA Serum Aldo Urine Aldo K hormone
Central diabetes High (can be normal if Nl/low High Low, Low Nl Nl Nl Nl
insipidus adequate water taken in) rises after
dose of
AVP
Nephrogenic diabetes High (can be normal if Nl/low High Low, does High Nl Nl Nl Nl
insipidus adequate water taken in) not rise
after dose
of AVP
Psychogenic polydipsia Low/normal Nl Low/normal Low Low Nl Nl Nl Nl Nl
SIADH Low Nl-high Low High High Nl Nl Nl Nl High
Hyper-aldosteronism Nl with hypertension Nl-low Nl Nl Nl Low High High Low High
Hypo-aldosteronism Low High Nl Nl Nl High Low Low High
Hyporeninemic Nl High Nl Nl Nl Nl-low Low Low High
hypo-aldosteronism
Pseudohypo-­ Low High Nl Nl Nl High High High High
aldosteronism
Cerebral salt wasting Low High Low High Nl High High High low High
Glucocorticoid Low Low Low High Nl (high Nl-high Depends on Depends on Low-Nl Low
insufficiency (dilutional in some cause of ad-renal cause of adrenal
hyponatremia) studies) insufficiency insufficiency
Hypothyroidism Low Low Low High Nl Low Low
(dilutional
hyponatremia)
Hyponatremia due to Low (1.6 mEq/mL Osmotic High High High
hyperglycemia decrease per 100 mg/dL diuresis
elevation of glucose)
Partially based upon table from Quest Diagnostics/Nichols Institute and sources in Suggested Readings
AVP arginine vasopressin, PRA plasma renin activity, Aldo aldosterone, Nl normal, SIADH syndrome of inappropriate secretion of antidiuretic hormone
4  The Posterior Pituitary Gland and Disorders of Vasopressin Metabolism
Diabetes Insipidus 35

tumor or anatomic abnormality should be carried


out even in young cases, unless a clear family pat-
tern or gene diagnosis is made. All of these condi-
tions are detailed in other chapters.

Nephrogenic Diabetes Insipidus

High transection If the defect is in the nephron, ADH is produced in


Low transection normal or increased amounts but is ineffective in
Anterior
controlling the production of urine. Originally,
pituitary Posterior
nephrogenic DI (NDI) was thought to be solely an
pituitary X-linked disorder, but numerous cases of sporadic
occurrence or autosomal dominant inheritance
Fig. 4.2  Level of transection of the pituitary stalk and have since been described. Thus congenital neph-
central diabetes insipidus. If a transection of the pituitary rogenic DI (NDI) may be the result of an X-linked
stalk is high, the axons of all vasopressin-containing neu- mutation in the renal vasopressin receptor (V2) at
rons are likely to be cut and to degenerate, leading to cen-
tral diabetes insipidus. If the transection is low on the Xq28 (*304800 DIABETES INSIPIDUS,
pituitary stalk, sufficient vasopressin-containing neurons NEPHROGENIC, X-LINKED), type 1 NDI. In
are likely to remain to avoid central diabetes insipidus this condition, a defect in the G protein of the
adenyl cyclase system of the renal tubular cells
leads to inability of vasopressin to increase uri-
p­ ituitary deficiencies often appeared and growth nary levels of cyclic AMP. This is the cause of
hormone deficiency was the most common condi- 90 % of congenital NDI cases. Alternatively an
tion that developed. The authors recommended autosomal recessive or autosomal dominant muta-
monitoring the pituitary stalk every 6 months for tion in the renal water channel aquaporin-1 at
3 years to see if further changes in thickness 12q13 (#125800 DIABETES INSIPIDUS,
occurred but found no further changes occurred NEPHROGENIC, AUTOSOMAL DOMINANT),
after 3 years. Congenital midline defects of the type 2 NDI leads to type 2 NDI. In this condition,
CNS also may lead to DI in addition to anterior vasopressin does cause increased urinary levels of
pituitary deficiencies (see Chap. 3). Absence of cAMP but urine excretion remains excessive.
the septum pellucidum may be associated with These are the remaining 10 % of cases.
optic hypoplasia or dysplasia and is often associ- Because the disorder classically is found as a
ated with hypothalamic abnormalities (#182230 congenital condition, these babies are more prone to
ICD+ SEPTOOPTIC DYSPLASIA at 3p21.2- dehydration than are the majority of patients with
p21.1); initial clinical presentation may be a central DI who have DI as an acquired defect.
visual deficiency (with to-and-fro or pendular Severe dehydration due to fluid loss may cause epi-
nystagmus due to the defect in the optic nerve) or sodes of unexplained fevers, vomiting, failure to
an endocrine disorder, including DI. The Wolfram thrive, and growth failure and even lead to develop-
(#222300 ICD+ WOLFRAM SYNDROME 1; mental delay; hypernatremia may be demonstrated
WFSOMIM) also called DIDMOAD (DI, diabe- during the episodes of dehydration. Intracerebral
tes mellitus, optic atrophy, and deafness) syn- calcifications of the frontal lobes and basal ganglia
drome is caused by a mutation of a gene at 4p16.1. may be noted after repeated episodes of dehydra-
Although histiocytosis X and craniopharyngio- tion. Nonobstructive ­hydronephrosis, hydroureter,
mas usually occur at a later age, and the early and megabladder may develop due to massive pro-
onset of anterior and posterior pituitary disorders duction and flow of urine in untreated cases.
usually carries a more benign prognosis than does Usually, when the child reaches an age at
late onset, MRI evaluation for a potential CNS which water can be obtained ad lib, the symptoms
36 4  The Posterior Pituitary Gland and Disorders of Vasopressin Metabolism

will decrease except at time of illnesses, when Table 4.2  Water-deprivation test
debilitation may cause a decrease in oral intake. A water-deprivation test can be very difficult for patient
Acquired nephrogenic DI may be due to drugs and the family. The child will be uncomfortable in
many cases and cry for fluids which of course would
such as lithium chloride and demeclocycline or
negate the test if administered. There is also danger of
to electrolyte abnormalities such as hypercalce- dehydration if the child has true diabetes insipidus, so
mia or hypokalemia, all of which affect the abil- careful observation is mandatory.
ity to concentrate the urine. Determine if patient is taking medications that can
cause polyuria or has diabetes mellitus or other chronic
disease that can cause decreased urine concentration
(see text). Does patient have apparent psychological
Clinical Features of Diabetes Insipidus condition leading to psychogenic polydipsia? If not,
proceed.
Clinical features of DI of either the central or nephro- Normal overnight fast: if the patient does not normally
genic variety relate mostly to patterns of drinking and drink overnight, fast, but if the patient does need to
drink, allow this, but document time of intake and
urinating. The patient will urinate large quantities of amount.
dilute urine throughout the day and night, awakening Determine first a.m. urine, serum osmolality, and
several times every night while constantly drinking hematocrit; if urine osmolality >600 (some have stated
(usually drinking cool water, as other fluids are less 450) mOsm, with serum osmolality ≤300 mOsm,
requested, at least in the US experience) because of vasopressin function is usually adequate.
continuous thirst. As noted, except for congenital If urine and serum do not match these guidelines, start
observed fast (some suggest giving normal breakfast and
defects, DI usually is first seen after infancy. If infants then start observed fast, but if patient is stable and fasted
are affected, they will cry if deprived of their bottle overnight, this may be omitted to shorten the test).
and may drink their bath water or suck on their wash- Check weight and urine osmolality hourly, and keep
cloths; these symptoms may be more common in track of urine volume. Measure serum sodium and
those with nephrogenic DI than in those with the cen- osmolality hourly, and measure serum vasopressin at 0
time and the end of the test (certainly before any
tral form. Older children and adults will go to bed vasopressin is given) for future reference. All tests
with liters of water at their side for use throughout other than vasopressin should be ordered stat.
the night. At times of disability or when water can- If urine osmolality >600 mOsm (some say 450) with
not be obtained, the patient with DI will become serum osmolality ≤300 mOsm, or if urine osmolality
>1,000 mOsm at any serum osmolality, function is
severely dehydrated and could develop shock with
normal.
high serum sodium values. Remarkably increased
If by 8 h after beginning of observed fast, urine
urine flow, virtually clear in color, into the diaper of osmolality <600 (some say 450) mOsm, or at any time,
an infant or enuresis in an older child may be seen. if serum osmolality is >300 mOsm/L and urine
Because of the high flow of urine, the renal pelvises osmolality stays below the guidelines, give 1 U/m2 of
aqueous vasopressin. If urine osmolality doubles the
and ureters may be dilated on intravenous pyelogram
previous value over the next hour, the diagnosis is
as secondary complications. These features are in central DI. If no change occurs, the diagnosis is
contradistinction to those found in the compulsive nephrogenic DI or some variant.
water drinker, who usually will get through the night If blood pressure drops, pulse rises abnormally, or if
without awakening, and the child drinking for second- weight drops >5 %, consider stopping the test and
obtaining last samples as listed. If the patient appears
ary gain, who drinks small amounts frequently and clinically stable, the test may be continued under
who urinates frequently but in small quantities (psy- careful scrutiny.
chogenic polydipsia). DI diabetes insipidus

Diagnosis of Diabetes Insipidus diabetes mellitus, chronic renal disease, or urinary


tract infection that explains the symptoms
Before proceeding with any detailed evaluation (Table  4.2; Fig. 4.3). A urine sample should be
for DI, it should be established that a patient free of sugar and be dilute compared with plasma
who has polyuria and polydipsia does not have osmolality to suggest diabetes insipidus. The first
Diabetes Insipidus 37

History reveals large volume of urine and frequent episodes of drinking during night and day, not frequent episodes of drinking small volumes
Yes No

immediately test urine for sugar and ketones possibly psychogenic polydipsia or urinary tract infection
negative positive

test first AM urine evaluate for diabetes mellitus


by serum glucose

concentration < 1.010 concentrated over 1.010

perform water deprivation test yes

can't concentrate urine concentrates urine


after deprivation

eliminates classic diabetes insipid,


exogenous vasopressin partial defect is possible

concentrates urine doesn't concentrate urine

central diabetes insipidus nephrogenic diabtes insipidus

Fig. 4.3  The diagnosis of polyuria/polydipsia. This is a be useful to ask the parents to keep a logbook of how much
general schema for the diagnosis of the child who is appar- the child drinks and how often the child urinates. If urinary
ently urinating too much. Before any evaluation, it would volume could be measured, it would be even more useful

question of importance is whether the patient of osmolality is preferable). Serum osmolality


really is ­urinating frequently in large quantities and sodium should be determined; in DI, the val-
through the day and night or whether it is simply ues will be normal or slightly elevated if the
a pattern of frequent but small episodes of urina- patient has free access to water and a normal thirst
tion. A diary of intake and output should be con- mechanism, whereas in psychogenic polydipsia,
structed, and determination of the 24-h urine the values will be normal or even low because of
volume at home under the watch of reliable par- the dilutional effect of excessive water drinking.
ents should be done to determine if this informa- An ambulatory, conscious patient without a thirst
tion supports a pattern suggestive of DI. If mechanism will often have a high osmolality and
urination occurs during the day while the child is sodium because such patients do not have the nor-
awake and not at night and there is no enuresis, mal drive to increase water intake as their serum
diabetes insipidus becomes much less likely. The sodium becomes increasingly concentrated; this
history should support a pattern suggestive of DI, condition has been called primary hypernatremia
and observation in the hospital or under the watch in some reports.
of reliable parents should be the first step to con- Urine volume greater than 4 mL/kg/h and
firming increased urine volume output and fluid plasma osmolality over 300 mOsm/L and serum
intake. The first voided urine sample in the morn- sodium over 145 Eq/L is compatible with a diag-
ing is normally the most concentrated of the nosis of diabetes insipidus. Since a conscious
whole day, and a sample should be subjected to patient with a thirst mechanism can keep up with
analysis of specific gravity and osmolality to see the sodium loss, the serum sodium and serum
if this sample is concentrated more than osmolality do not necessarily have to be elevated.
750 mOsm/L, which would eliminate the diagno- If the tests noted still suggest that the patient
sis in most cases (specific gravity may be has DI, a careful water-deprivation test should be
increased fallaciously by contamination with non- performed. If not carried out appropriately, there
osmotic substances, e.g., stool, so measurement is danger of severe dehydration with such a test.
38 4  The Posterior Pituitary Gland and Disorders of Vasopressin Metabolism

These tests are difficult for the child and perhaps and urine osmolality should be compared and a
even more so for the family. The child will be serum vasopressin determination obtained to match
expected to complain about lack of water whether with the osmolality determination (the vasopressin
DI or psychogenic polydipsia is involved, and the value will not return for weeks from the laboratory
parents likewise might be uncomfortable over the but may prove useful if the diagnosis is still in
procedure as the child cries for water. The most doubt later).
important consideration of such a test is to As a patient becomes dehydrated, the urine
observe the child constantly for signs of dehydra- osmolality will increase because of the delivery
tion that may be dangerous or for “cheating” of increasingly concentrated filtrate to the kidney,
when the child takes or gets water surreptitiously, but a patient with DI cannot concentrate the urine
which will nullify the test results. The test should to more than 1.5–2 times the serum osmolality.
be done only with full staffing, and the later part If the serum osmolality increases to 300 mOsm/L
of the thirst phase of the test should not occur at or higher, the urine concentration should nor-
night if staffing is low and the child might not be mally be more than 450 mOsm/L and ideally
well observed. more than 600–750 mOsm/L. In an intermediate
The child should have a normal dinner, and situation in which a trend toward dehydration is
the usual nighttime routine should continue. developing, a continuation of the fast may be
If the child stays at home for this night, whatever necessary; this will be safe only if careful obser-
is the usual routine is followed (no fluid intake, if vation of state of hydration, BP, and pulse is con-
the child has been able to tolerate this schedule tinued. Further, in partial DI, a patient may pass
in the past, or fluid intake, if that is the norm). one test with just adequate urinary concentration,
The next morning, the first voided urine should be whereas, if the test is repeated the next day, the
analyzed for osmolality, the body weight and BP patient may be virtually unable to concentrate the
should be determined, and serum sodium, osmo- urine because of the exhaustion of the patient’s
lality, and hematocrit should be determined. meager supply of vasopressin.
Assuming that the urine is not concentrated to If the serum osmolality has increased without
600–750 mOsm/L or greater and the serum concentration of the urine and therefore no
osmolality is less than 300 mOsm/L (reaching increase in urine osmolality, 0.05–0.15 mL of
these values demonstrates normal vasopressin d-arginine-d-amino-vasopressin (DDAVP, see
physiology), the test may commence. At that later for explanation of this medication) in a nos-
time, all oral intake should cease if it had not tril (or, if DDAVP is unavailable or there is
ceased during the night. Weight and BP should be pathology of the nasal mucosa, a 1-U/m2 subcuta-
taken hourly, serum osmolality and hematocrit neous dose of aqueous vasopressin) is adminis-
determined every 2 h, and all urine volume moni- tered, and the volume of urine and the urine
tored and osmolality measured at every void. concentration in the next 30–60 min is compared
The test should cease if the BP decreases 10 %, with the values obtained before the DDAVP or
or the serum osmolality increases above exogenous vasopressin was given. In central DI,
300 mOsm/L. If the urine osmolality increases at the end of the fast, the patient has secreted the
above 600–750 mOsm/L but the serum osmolal- maximal vasopressin that he or she is capable of
ity is not above 300 mOsm/L, the test should secreting, but this diminished store of vasopres-
stop, as there is no likelihood of DI. If the serum sin is inadequate for the task of concentrating the
osmolality does not rise above 300 mOsm/L and urine. Thus, the exogenous vasopressin will fur-
the urine osmolality does not rise above ther concentrate the urine twofold. A patient with
450 mOsm/L, the water-deprivation test should nephrogenic DI will not be able to concentrate
continue as the next few hours may lead to the urine past 450 mOsm/L in spite of increasing
diagnosis. Otherwise, at the end of 8 h or up to serum osmolality, and the addition of exoge­
10 h, if adequate staffing is available, the serum nous vasopressin will not further increase urine
Diabetes Insipidus 39

o­ smolality or reduce urine volume, because the  reatment of Central Diabetes


T
patient is resistant to vasopressin. Insipidus
The diagnosis of psychogenic polydipsia may
sometimes present problems. Because of the An untreated patient with DI, if old enough and
patient’s excessive drinking habits and the excess able enough to take in sufficient water, will
water load chronically presented to the kidney, likely survive but will develop dilated ureters
the patient may have developed a dilute or and dilated renal pelvises because of the
“washed out” medullary gradient, leading to a increased urinary flow and will be on the brink
situation which might be considered similar to of severe dehydration if anything disturbs the
moderate nephrogenic diabetes insipidus. If this balance achieved (see Chap. 14 for description
is owing to a long history of excess water intake, of emergency treatment of hypernatremia due
the patient may not reach full urinary concentra- to DI). The appropriate therapy is vasopressin
tion even if the fast is continued and if endoge- replacement in a convenient form. Native vaso-
nous vasopressin is adequate. However, pressin has a short half-life and exerts hyperten-
exogenous vasopressin at the end of the water- sive effects. Desamino-d-arginine vasopressin,
deprivation test will not cause further concentra- or desmopressin or DDAVP, an altered vaso-
tion of the urine because in a patient with psycho- pressin molecule that has 140 times the urine-­
genic polydipsia exogenous vasopressin exerts concentrating ability but almost none of the
no further effect than the endogenous vasopres- vasoactive effects of native arginine vasopres-
sin. Usually, the psychological history in addition sin, is the treatment of choice. A dose of DDAVP
to the results of the water-deprivation test will will usually last approximately 12 h (or even
lead to the correct diagnosis, as most children 24 h), whereas native vasopressin has a half-life
with psychogenic polydipsia will not have such a of 20 min. Oral DDAVP tablets are available in
degree of urinary dilution after a fast as will those doses of 01 and 0.1 mg given 1 to 3 times per
with nephrogenic DI. day. The dose of oral DDAVP tablets is titrated
Because central DI is possibly due to a con- until adequate antidiuresis is achieved after a
genital anatomic defect of the CNS or an acquired nighttime administration. A second morning
CNS tumor, an MRI is indicated. Loss of the T1-­ dose is given if significant polyuria still occurs
weighted bright spot of the posterior pituitary during the day and a third dose may be needed
gland during gadolinium MRI scan will suggest as well in some. Alternatively, DDAVP is
central DI associated with the loss of vasopressin administered in measured doses through inhala-
and neurophysin production that are responsible tion by the nostrils. A dose of 0.025 mL of a
for the hot spot. However, in nephrogenic DI, a solution of 10 μg/0.1 mL (yielding 2.5 μg) is
loss of the bright spot may be due to continued given with a plastic tube inserted into the nostril
increased release of vasopressin. The changes and is repeated when the child complains of
in the bright spot may be found in normal indi- increasing thirst or an infant begins to increase
viduals as well as in situations of extreme thirst urinary frequency again. A syringe may be used
or overhydration and so is not highly specific. In to squirt DDAVP into the nostrils of the young-
congenital loss of pituitary function, the appear- est children. A measured dispenser of nasal
ance of an interrupted pituitary stalk or ectopic spray is another treatment, with a dose of 10 μg
bright spot might be indicative of the diagnosis. per spray (0.1-mL volume). Lysine vasopressin
In some cases, a thickening of the pituitary stalk or Diapid is given as a nasal spray (50 U/mL or
is all that suggests a developing tumor; sequential 2 U per spray) that lasts 2–8 h, if a short action
follow-up MRI is indicated, as it may take years is desired (e.g., in infants). This treatment is
for the tumor to be truly discernible. Tumor rarely used at present.
markers in the blood or CSF (e.g., hCG in germi- A patient must have a phase of urinary dilu-
nomas) should be monitored if a tumor is strongly tion or breakthrough urination before the next
suspected. dose of DDAVP is given to ensure that water
40 4  The Posterior Pituitary Gland and Disorders of Vasopressin Metabolism

intoxication does not result. Hyponatremic sei- equal to the output for the last 1–4 h above 40 mL/
zures are a concern if the patient does not have kg/h; replacing one-hour output volume over the
some urination during the day. In the presence of next hour of intravenous fluids is the best method.
an intact thirst mechanism, the patient should be A limited maximal fluid replacement might be set
able to maintain a normal sodium concentration at 120 mL/kg/h to avoid the development of
with DDAVP treatment. hyponatremia. Glucose may rise during this regi-
The treatment of young infants with diabetes men, and there are reports of exceptionally high
insipidus presents significant difficulty in maintain- blood glucose values due to volumes of replace-
ing stable serum sodium levels with the use of ment higher than recommended above with fluids
DDAVP. Some have been treated with breast milk containing more than 5 % dextrose.
or low solute formula (Similac 60/40) and water, Vasopressin administration in short-term prep-
but this has been associated with poor growth. The arations is an alternative treatment to full intrave-
use of hydrochlorothiazide in 5–10 mg/kg adminis- nous fluid replacement in patients with diabetes
tered two or three times per day to foster sodium insipidus. However, the administration of vaso-
retention leads to free water retention and a more pressin may cover up the development of diabetes
stable situation. Parental DDAVP is not yet approved insipidus in the triphasic response after pituitary
for infants but might offer a future therapy. surgery as noted below, so a high index of suspi-
Intravenous fluid management of hospitalized cion for diabetes insipidus must be upheld; fur-
patients with diabetes insipidus cannot be carried ther administration of excessive fluids while
out by the customary formula for maintenance vasopressin is released can lead to an SIADH-like
fluid management. A patient without diabetes picture. Intravenous (i.v.) vasopressin manage-
insipidus will in the basal state have about 60 % ment during surgery or recovery is 0.5–2 mU/
of their maintenance requirements due to urinary kg/h of vasopressin while carefully monitoring
output. The patient with diabetes insipidus will fluid intake to ensure appropriate fluid balance.
have more or less than that 60 % based upon hos- Intravenous vasopressin may stick to the intrave-
pital fluid management and exogenous vasopressin nous tubing, and a slightly higher dose may be
preparations rather than normal physiology. Thus required so the dose needs to be titrated to the uri-
the standard formulas do not apply. nary output in a manner rather similar to titrating
Patients with diabetes insipidus in the postop- intravenous insulin by measuring the change in
erative state in the intensive care unit for some blood sugar in diabetic ketoacidosis. When vaso-
other illness may be best managed without long-­ pressin is used in fluid management, it is recom-
acting vasopressin which may complicate fluid mended that total fluid administered over 24 h
management. A hospitalized patient not managed does not exceed 1 L/m2/day which is equivalent to
appropriately may very well be underhydrated two-thirds maintenance. Excessive fluid adminis-
leading to hypernatremia and then overhydrated tration with i.v. vasopressin or any of the forms of
leading to hyponatremia. Acute diabetes insipi- vasopressin listed earlier will lead to an SIADH-
dus that occurs intraoperatively during central like condition. If the patient is undergoing sur-
nervous system surgery or in the postoperative gery, a nursing order after surgery is frequently
state in a patient with chronic diabetes insipidus “push fluids”; if the patient has been treated with
can be carried out with intravenous fluids admin- a vasopressin dose when the increased “pushed”
istered according to the schema of output equals fluids are administered, an SIADH-like syn-
input with a small basal infusion maintained drome is a risk. The best way to lower the risk for
throughout even without vasopressin administra- overhydration in the child that has a normal thirst
tion. The patient will receive 40 mL/kg/h of mechanism is to switch to oral fluids as soon as is
appropriate maintenance fluid with extra fluid possible. Then taking the direction from the
added with electrolytes, to cover urinary volume child’s thirst mechanism will make excessive
output. Thus careful records of urinary output are fluid administration less likely.
kept, and intravenous fluid for urinary volume Patients with an absent thirst mechanism and
replacement is administered at 1–4-h intervals diabetes insipidus are extremely difficult to treat.
The Syndrome of Inappropriate Secretion of Antidiuretic Hormone 41

A set regimen is empirically determined under analogs called pharmacologic chaperones that
careful observation so that a given number of cross the plasma membrane and reach misfolded
glasses of water or a volume of water per day suf- AVPR2 receptors, helping them to refold and
ficient to keep the serum sodium in the normal reach the plasma membrane where endogenous
range is prescribed. An extra glass of water or vasopressin displaces the vasopressin analogue
more should be given for moderately increased and activates the receptor. Thus the mutant recep-
activity, exposure to high temperature, or illness tor is rescued by these “chaperones” which treat
especially with fevers. Measurement of urine the nephrogenic DI. Chaperones are not in clini-
output matched with administration of an equal cal use in children as yet.
volume of oral fluids with insensible loss added
to this will be the best way to maintain normal
osmolality. Every week or two, the serum sodium  he Syndrome of Inappropriate
T
should be measured, with determinations more Secretion of Antidiuretic Hormone
often if stability has not been reached. The seri-
ous consequences of dehydration or overhydra- In the presence of increased vasopressin secretion
tion should not be minimized in this complex or vasopressin effect and excessive water intake,
condition as serious illness or death may occur if hyponatremia will develop in the absence of the
not adequately treated. ability to excrete adequate fluid, and the condition
is known as the syndrome of inappropriate secre-
tion of antidiuretic hormone (SIADH used here)
 reatment of Nephrogenic
T or syndrome of inappropriate antidiuresis (SIAD).
Diabetes Insipidus (If the stimulus for the ­vasopressin secretion is
physiologic such as volume depletion, e.g., due to
The goal of treatment of nephrogenic DI is to diarrhea, the term syndrome of appropriate antidi-
cause the kidney to retain water in the absence of uretic hormone secretion (SAADH) has been sug-
any effects of vasopressin; the method may seem gested although is not widely used.)
paradoxical, as a low-sodium diet and diuretic Classic features of SIADH are (1) plasma
therapy is prescribed. With decreased serum sol- hypoosmolality (<275 mOsm/kg), (2) less than
ute due to the low sodium intake and the loss of maximally dilute urine (urine osmolality
sodium due to iatrogenic diuresis, the site of >100 mOsm/kg), (3) euvolemia (secondary to reg-
reabsorption of water shifts from the collecting ulatory adaptations after initial increased intravas-
duct where vasopressin exerts its effect to the cular volume), (4) natriuresis, (5) normal renal
proximal tubule where aldosterone, maximally function, and (6) no evidence of thyroxine or cor-
stimulated by whole-body sodium loss, will act tisol deficiency which would promote water reten-
to retain all available sodium and thereby carry tion. This constellation is paradoxical because the
water back into the vascular compartment along low serum sodium and osmolality could be cor-
with the sodium, thereby leading to decreased rected if the urine is maximally diluted and if max-
urine flow. Thus the treatment bypasses the imal urinary sodium reabsorption is accomplished.
tubule where vasopressin exerts effects to an (SAADH is found with these features except the
area where aldosterone exerts its effect. A thia- patient is hypovolemic.)
zide diuretic with the addition of amiloride (0.4– Because the combination of vasopressin
0.625 mg/kg/day) to counteract the hypokalemia secretion and water intake is responsible for
caused by thiazide is commonly employed. SIADH, the syndrome is often iatrogenic; the
Alternatively, thiazide diuretics and indometha- patient already may be under a physician’s care
cin (2 mg/kg/day) or amiloride are used, with a and may be kept on the same regimen of fluid
careful observation for nephrotoxicity. therapy after vasopressin secretion begins fol-
New therapy that may enter general use lowing an episode of temporary DI which has
involves cell permeable lipid-soluble vasopressin resolved or has been given excess fluids after
42 4  The Posterior Pituitary Gland and Disorders of Vasopressin Metabolism

vasopressin is administered as a treatment for Another condition combining hyponatremia


DI. Atrial natriuretic peptide may be responsible associated with various types of CNS disease is
for the sodium ­diuresis, as it is elevated in times cerebral salt wasting (CSW); CSW is described
of volume overload. If fluid therapy in the treat- in head injury, brain tumor, intracranial surgery,
ment of conditions predisposing to SIADH was craniosynostosis repair, stroke, intracerebral
appropriately regulated, and if serum sodium hemorrhage, and tuberculous meningitis. In this
concentrations were monitored routinely, condition, hyponatremia along with increased
SIADH would be far less likely. Patients with urinary sodium excretion which may exceed
cancers that produce vasopressin in ectopic 150 mEq/L, polyuria with decreased vasopressin
locations may be in the habit of drinking a set secretion, is characterized by hypovolemia in
amount of fluid before the tumor developed; if contrast to SIADH, in which intravascular volume
the same fluid intake is continued after the ecto- is normal or increased. In cerebral salt wasting,
pic vasopressin is present, SIADH may develop atrial natriuretic peptide concentrations are
even before the cancer itself is diagnosed. increased over 20 pmol/L and may be the primary
Any disorder of the lungs, including those defect although increase in CNP is also suggested
requiring ventilator support, can cause increased as an etiology. The values of serum aldosterone
release of vasopressin, mediated by the volume and vasopressin are normal or decreased in cere-
or stretch receptors in the right atrium. Thus, the bral salt wasting. Treatment involves the replace-
common prescription for increased fluid intake ment of sodium losses and volume and if possible,
during episodes of pneumonia or other disorders treatment of the underlying central nervous
of the lungs may precipitate an episode of ­system condition.
SIADH. Patients supported on ventilators for Determination of plasma volume is most
extended periods must have serum sodium regu- accurately carried out by central venous line. If
larly monitored. Infant botulism may lead to there is no central venous line, laboratory studies
SIADH during ventilator support as this may can be supportive of the determination. In hypo-
trigger increased vasopressin secretion and volemia, plasma renin, angiotensin II, aldoste-
SIADH. Most neurologic conditions, including rone, vasopressin, urinary sodium, and fractional
meningitis, tumors, postsurgical condition, and excretion of sodium are low. In hypervolemia,
trauma, can increase vasopressin secretion; urinary sodium and fractional excretion of
these potential complications are well recog- sodium are high, while urine osmolality is low.
nized and account for the usual orders for Fractional excretion of sodium is calculated as
reduced fluid administration in neurologic dis-
U Na ´ PCr
ease. However, after any surgery, increased FE Na = ´100
vasopressin secretion occurs, and the patient is PNa ´ U Cr

susceptible to SIADH. Any condition causing
nausea and emesis, including carcinomatosis or It must be stated that there is controversy as to
the administration of chemotherapy, can the existence and prevalence of cerebral salt
increase vasopressin secretion. Further, drugs wasting in the literature with a critical assessment
often used in cancer therapy, such as vancomy- suggesting that plasma volume is rarely accu-
cin, vincristine, and cyclophosphamide rately determined in states of hyponatremia.
(Cytoxan), increase vasopressin secretion in
addition to the increased vasopressin secretion
due to nausea. Many types of cancers produce Nephrogenic Syndrome
vasopressin in an ectopic hormone-­ secreting of Inappropriate Antidiuresis
syndrome, so that a patient with cancer may be
susceptible to SIADH from the cancer, from the Recently, the nephrogenic syndrome of inap­
nausea of the cancer therapy, and from the che- propriate antidiuresis (NSIAD) (#300539
motherapeutic agent itself. NEPHROGENIC SYNDROME OF
The Treatment of Syndrome of Inappropriate Secretion of Antidiuretic Hormone 43

INAPPROPRIATE ANTIDIURESIS; NSIAD) elevation of blood glucose of 100 mg/dL causes a


was described as a consequence of a gain-of-­ decrease in serum sodium by 1.6 mEq/mL.
function mutations in the gene encoding the vaso- Low serum sodium or pseudohyponatremia
pressin V2 receptor (AVPR2; 300538). These will result from hyperlipidemia and hyperpro-
patients have what clinically appears to be classic teinemia (such as in multiple myelosis). Of
SIADH but without measurable AVP values. course, if the sodium sample was taken from an
intravenous line or downstream from an intrave-
nous line, the sample will be composed of
Other Causes of Hyponatremia ­hypotonic fluid, and the serum sodium will fal-
laciously decrease in the laboratory determina-
Not all episodes of hyponatremia are due to tion. Pseudohyponatremia is asymptomatic and
SIADH. The most frequent cause of hyponatre- is not a physiologic problem, as the cells are actu-
mia in pediatrics is fluid overload with hypo- ally exposed to normal amounts of sodium in
tonic fluids while receiving i.v. therapy under a spite of abnormal sodium on laboratory determi-
physician’s care. Indeed some authorities rec- nations. Thus in pseudohyponatremia, correction
ommend avoidance of hypotonic fluids for of the underlying condition is indicated, but
maintenance or repair in most situations and sodium administration is rarely necessary.
instead to use isotonic fluids. However, the basis
of pediatric fluid management, as developed
more than 50 years ago, recommends isotonic  he Treatment of Syndrome
T
fluid for repair of fluid deficit but consideration of Inappropriate Secretion
of lower tonicity fluids for maintenance. These of Antidiuretic Hormone
controversies cannot be addressed in this chap-
ter except to reemphasize the basics of fluid The treatment of SIADH is first and foremost
management as noted. prevention (see Chap. 14 for discussion of emer-
In addition, congestive heart failure and the gency treatment of hypernatremia due to SIADH).
oliguric phase of acute renal failure may lead to This is often an iatrogenic disease, and monitor-
hyponatremia due to inappropriate fluid adminis- ing fluid therapy and frequent serum sodium
tration in the face of decreased urinary output. concentration determinations will lessen the
­
Total body sodium is depleted in diabetic keto- possibility of severe shifts of sodium. Once
­
acidosis as described in Chaps. 11 and 14. SIADH develops, fluid restriction to the ­minimum
While glucose does not normally play a sig- possible but safe level is the first approach.
nificant role in serum osmolality, when glucose In many cases, maintaining i.v. fluids at a level
cannot enter cells through insulin-dependent glu- just able to keep the i.v. line open is appropriate
cose transporters due to insulin deficiency or for a time as the attainment of fluid balance is
resistance, glucose becomes an important osmot- accomplished. With less severe hyponatremia,
ically active ion when hyperglycemia develops. replacing urine output with an equal volume of
Thus osmolality is higher in the extracellular i.v. fluid calculated every 2–4 h is adequate fluid
space then the intracellular space which will trig- replacement. Because the urine flow is so low in
ger vasopressin secretion. SIADH, the tendency will be to fear that inade-
An elevation of atrial natriuretic factor occurs quate intravascular volume is the problem, and
in some cases of diabetic ketoacidosis which is a it may appear that administration of boluses of
condition which causes true hyponatremia requir- fluid to increase urinary output is appropriate.
ing replacement therapy. Fluid retention will If the oliguria is due to dehydration, the boluses
lower serum sodium adding to the decrease in are appropriate and will allow urination, but if
serum sodium due to sodium depletion. SIADH has been diagnosed, the boluses will only
Measurement of serum sodium can be corrected intensify the SIADH, as the fluid will be retained.
for the hyperglycemic effect of glucose; each Careful review of the records, including an
44 4  The Posterior Pituitary Gland and Disorders of Vasopressin Metabolism

accounting of all fluids administered (a balance the use of hypertonic saline or the combination of
sheet of intake and output is essential), changes furosemide (Lasix) and hypertonic saline described
in body weight, and an estimation of intravascular earlier may be the only appropriate ­ therapy.
volume or measurement of central venous pressure Whatever therapy is offered, constant observation
should clarify the diagnosis. must be given for severe fluid shifts.
Other methods are available to break through Rate of correction of SIADH is of importance.
the SIADH and allow urine flow so that intravas- If the hyponatremia of SIADH lasted more than
cular volume decreases and the condition is mod- 48 h, correction of hyponatremia too rapidly can
ified. Lithium and demeclocycline will interfere lead to central pontine myelinolysis which will
with ADH action on the kidney causing iatro- develop in 24–48 h and may be diagnosed on
genic NDI, but the inherent side effects limit their MRI. It is recommended to correct serum sodium
use (especially demeclocycline) in younger chil- to 120–125 mEq/L at a rate of no greater than
dren. Urea has been used as an osmotic diuretic 0.5 mEq/L/h, with no more than 12 mEq/L cor-
in pediatric SIADH and NSIAD. Clinical rection occurring in the initial 24 h and 18 mEq/L
research studies using nonpeptide V2R antago- in the initial 48 h of treatment.
nists (known as vaptans) progress in children but
are not in general use in pediatrics although some
vaptans have been approved by the Food and  he Triphasic Response After
T
Drug Administration for use in adults. Surgery for Craniopharyngioma
Replacement of the volume loss with 3 % saline
will help correct symptomatic hyponatremia A patient with a craniopharyngioma or other
without administering excessive fluid volume to tumors of the hypothalamus or posterior pituitary
worsen the problem. The administration of area already may have DI before surgery is car-
12 mL/kg of 3 % saline will cause a 10 mEq/L ried out for the tumor; if not, when the pituitary
rise in serum sodium. If hyponatremia is severe stalk is cut, DI may manifest immediately, pos-
and seizures have resulted, furosemide may be sibly while the patient is in the operating suite
administered to decrease serum volume along (Fig. 4.4). After this, in a few days, often unre-
with 3 % saline which is administered in a vol- strained release of vasopressin will occur as the
ume smaller than the diuresis caused by furose- cut nerve cells degenerate. If the high level of
mide, to bring about a net increase in serum fluid replacement originally necessary for treat-
sodium values thus fostering urination without ment of the DI is continued during this secondary
inappropriately increasing volume. This proce- phase of vasopressin secretion, SIADH will
dure of diuretic and hypertonic fluid administra- develop. During the following days, a third phase
tion can cause rapid fluid shifts, causing dilution of permanent DI will occur in most patients.
and concentration at various times, and is poten- However, some patients who receive the surgical
tially dangerous. Careful monitoring and main- transection low on the pituitary stalk will resume
taining a patent i.v. line to administer fluid adequate vasopressin secretion for needs, and the
volume if necessary are essential. DI will cease. It is imperative that fluid output be
It should be emphasized that the sodium diure- monitored carefully after pituitary stalk surgery
sis characteristic of SIADH is continuous during so that intake is matched to output, and fluid
the active phase of the disorder, and sodium admin- overloading does not occur during these chang-
istered by any route will quickly be passed out in ing fluid dynamics, or that dehydration does not
the urine and will not offer a long-­term cure of occur. This triphasic response occurs in about a
hyponatremia. However, if seizures are intractable, third of patients having such surgery.
Suggested Readings 45

More concentrated
High volume
Phase 3 of DI in most

Urine
Urine Flow
Concentration

and Phase 2 of and


Phase 1 of DI ++++++++++
SIADH
Normal +++++ Normal
+ ++++
++ Phase 3

+
+++
Reverts to normal
in some

Low volume
Less concentrated
1 2 3 4 5 2 3 4 5
Hours Days

Fig. 4.4  The triphasic pattern of urine concentration after release their content of arginine vasopressin leads to highly
pituitary surgery, usually for a craniopharyngioma. On the ­concentrated urine and decreased urinary excretion. This is
left Y-axis is urine concentration relative to a conceptional a period in which the syndrome of inappropriate ADH secre-
normal value and on the right axis is urine flow relative to a tion may develop especially if fluid volume is maintained at
conceptional ­normal flow volume. Immediately following a high level as was necessary in phase I. If the transection is
pituitary–hypothalamic surgery, the patient will have an low on the pituitary stalk, normalization of urine flow
inability to concentrate urine and large volumes of output (++++++) and urine concentration may occur (........) in the
results. The second phase when damaged neurons may third phase, rather than permanent diabetes insipidus

SIADH. Eur J Endocrinol. 2014. doi:10.1530/


Suggested Readings EJE-14-0530.
5. Moritz ML, Ayus JC. Intravenous fluid management
1. Majzoub JA, Muglia LJ, Srivatsa A. Chapter 11— for the acutely ill child. Curr Opin Pediatr. 2011;
Disorders of the posterior pituitary. In: Sperling MA, 23(2):186–93.
editor. Pediatric endocrinology. 4th ed. Philadelphia, 6. Wang J, Xu E, Xiao Y. Isotonic versus hypotonic
PA: Elsevier; 2014. p. 405–443. maintenance IV fluids in hospitalized children: a
2. Di Iorgi N, Allegri AE, Napoli F, Calcagno A, meta-analysis. Pediatrics. 2014;133(1):105–13.
Calandra E, Fratangeli N, Vannati M, Rossi A, 7. Foster BA, Tom D, Hill V. Hypotonic versus isotonic
Bagnasco F, Haupt R, Maghnie M. Central diabetes fluids in hospitalized children: a systematic review and
insipidus in children and young adults: etiological meta-analysis. J Pediatr. 2014;165(1):163–169.e162.
diagnosis and long-term outcome of idiopathic 8. Maghnie M, Cosi G, Genovese E, Manca-Bitti ML,
cases. J Clin Endocrinol Metab. 2014;99(4): Cohen A, Zecca S, Tinelli C, Gallucci M, Bernasconi
1264–72. S, Boscherini B, Severi F, Arico M. Central diabetes
3. Hannon MJ, Behan LA, O’Brien MM, Tormey W, insipidus in children and young adults. N Engl J Med.
Ball SG, Javadpour M, Sherlock M, Thompson CJ. 2000;343(14):998–1007.
Hyponatremia following mild/moderate subarach- 9. Rivkees SA. Differentiating appropriate antidiuretic
noid hemorrhage is due to SIAD and glucocorticoid hormone secretion, inappropriate antidiuretic hor-
deficiency and not cerebral salt wasting. J Clin mone secretion and cerebral salt wasting: the com-
Endocrinol Metab. 2014;99(1):291–8. mon, uncommon, and misnamed. Curr Opin Pediatr.
4. Matsuyama J, Ikeda H, Satoh S, Yamamoto K, 2008;20(4):448–52.
Ohashi G, Watanabe K. Early water intake restric- 10. Bichet DG. Clinical manifestations and causes of

tion to prevent the inappropriate antidiuretic hor- nephrogenic diabetes insipidus. http://www.uptodate.
mone secretion following transsphenoidal surgery; com/contents/clinical-manifestations-and-causes-of-
low body mass index predicts postoperative nephrogenic-diabetes-insipidus.
Disorders of Growth
5

Short stature is one of the most common complaints chart reflecting a decrease in the growth rate cannot
that bring a child to a pediatric endocrinologist, be assessed until another visit occurs, because
either by parental choice or by referral from the fam- two height measurements at least 3 months apart
ily’s physician. Growth can be considered a bioassay (and optimally longer) are needed to determine
of the state of health of the child as a normal growth growth rate accurately. If previous accurate
rate usually is a good indicator of health. Short stat- height measurements from well-child visits were
ure, rather than universally indicating an endocrine available, any change in growth rate would be
abnormality, can herald the onset of nonendocrine noted earlier. Because many systemic diseases
systemic disease, a state of malnutrition, or simply a affect growth, a decrease in growth rate, which
variation of normal. Since the goal of this chapter is would serve as an early indication of the onset of
the evaluation of endocrine disorders that affect such a disease, would be missed. Incorrect mea-
growth, other chronic conditions should be ruled out surements are responsible for numerous inappro-
before embarking on a sophisticated endocrine eval- priate referrals for short stature. Further, incorrect
uation. Thus initial history and physical examination measurement can obscure the effects of a medi-
should determine whether a search for endocrine cation meant to correct an abnormality of stature
conditions is warranted. It is especially important to (e.g., the positive effects of growth hormone
determine whether a nutritional condition has caused (GH) treatment can be evaluated only with accu-
decreased growth rather than starting an endocrine rate, sequential measurements) (Fig. 5.1).
evaluation. Tall stature or increased growth rate may An accurate measurement of infant length
also indicate an important condition. always requires two adults. The child must be laid
on a flat surface with a device that has one plate
horizontal to the plane of the top of the child’s
Measurement of Growth head and another plate horizontal to the first in the
plane of the child’s feet. The two plates should be
Measurement of stature is the cheapest procedure at a 90° angle to a ruler on which the child’s
available in the pediatric office and the one often height is read. Several available devices range
incorrectly performed if even performed at all! from inexpensive portable plastic caliper-like
Failing to measure a child at a visit is a serious devices (e.g., infantometer) to scales with the
mistake that limits the assessment of the health of measuring arms permanently attached (Fig. 5.2).
the child. Further, if a growth deficiency is devel- The worst method of measurement of infants
oping as a consequence of the medical condition is all too frequently in common use; a single
that precipitated the office visit, the most impor- observer makes a mark on the paper covering the
tant measure of an inflection point in the growth examining table at the foot of the child and

© Springer International Publishing Switzerland 2016 47


D.M. Styne, Pediatric Endocrinology, DOI 10.1007/978-3-319-18371-8_5
48 5 Disorders of Growth

a 5000 b
60

+250
55 +250
4000

Length, crown-heel (cm)


Mean
Mean 50
Birthweight (g)

3000 –250
–250 45

2000
40

1000
35

0 30
24 28 32 36 40 44 24 28 32 36 40 44
Week of gestation Week of gestation

c 40

36 +250
Mean
–250
32
Head (cm)

28

24

20
24 28 32 36 40 44
Week of gestation

Fig. 5.1 Intrauterine growth charts showing the normal growth of live-born Caucasian infants at sea level:
values of body weight (a), length (b), and head circumfer- Standards obtained from measurements in seven dimen-
ence (c) for infants born at different gestational ages at sea sions of infants born between 25 and 44 weeks of
level (Montreal) (from Usher R, McLean FM. Intrauterine gestation. J Pediatr 1969;74:901, with permission)

Sliding plate for the head


perpendicular to the board

Fixed plate for the feet perpendicular to the board

Fig. 5.2 A commercially available device used for mea- a movable perpendicular plate for the infant’s feet. It takes
suring lying length in infants. There is a ruler in centime- two adults to measure an infant after placing the infant
ters fixed to the horizontal board. There is a fixed straight on its back in the device
horizontal plate where the infant’s head is to be placed and
Measurement of Growth 49

another mark at the head to measure the distance


between the marks as an indication of the length
of the child. It should be obvious that the paper is
so crumpled by the weight of the child as to make
the distance between the head and feet quite vari- Horizontal plate that slides vertically
and is always perpendicular to the
able and that the movement of even a relatively backboard and is always parallel to the floor
quiet child will make such measurements useless.
It may be glib to say so, but guessing the length
of a term newborn as 21 in. is usually more accu-
rate than using the paper technique described for
a newborn!
The measurement of a patient older than 2
years is done with the child standing. The switch
from lying to standing measurements is respon-
sible for a large number of inappropriate referrals
because of the 1–2-cm decrease in height mea-
surements that occurs when switching positions.
It is important that the position of measurement
be indicated on the chart next to the numeric
measurement for children at this age of transi- Ruler in centimeters fixed to the
backboard
tion, so that unfounded worry about a declining
growth rate does not develop.
The device used to measure standing height
must be a variation of a standard stadiometer (e.g., Child’s back is firmly pressed to the upright
Harpenden stadiometer), which can measure to section. Child stands barefoot on flat surface
upon which the ruler abuts
0.1 cm. The child must have their back straight
against the wall or against a hard, flat surface on Fig. 5.3 A stadiometer allows the accurate measurement
which the stadiometer is attached. The device of standing height to an accuracy of a few tenths of a cen-
must be checked with a standard measure, often timeter. There is an indicator on the sliding plate which is
perpendicular to the back plate that indicates the height.
60 cm long, as measurements will vary over time There are mechanical and electronic versions of this
unless recalibrated daily. The child’s back must be device. A standard bar (usually of 2 m in length) is used to
straight and the heels and back must press calibrate the measurement daily. The child’s shoulders,
posteriorly to the surface. The feet must be on a buttocks, and heels are firmly pressed to the back plate
while the child stands barefoot on a flat surface which is
floor or hard surface, not on carpet, as the bottoms perpendicular to the back plate with the ruler. The child is
of the feet are considered to be the lower limit of to look straight ahead holding the head in the Frankfort
the measurement. The top of the measurement plane to standardize the position for the measurement
must be a plate completely parallel to the plane of with the top of the head parallel to the upper plate. The
examiner can gently hold the child’s jaw in position,
the feet, and the measurement must be read off a insuring that there is no slouching. Hair barrettes or other
stationary ruler that is at right angles to the planes ornaments must be removed. Unfortunately, if there is sig-
at the feet and head. The head is gently supported nificant braiding of the hair, it will be more difficult to
at the angle of the jaw with the child looking for- make an accurate measurement especially if the braiding
is not present on the next measurement as there is no way
ward horizontally into the distance (Frankfort to compare the two measurements
plane) to make sure that the head is horizontal to
the feet. A Harpenden stadiometer or an electronic
stadiometer is the most accurate of such devices, correct measurement procedures are followed
as it indicates the height in millimeter increments, (Fig. 5.3). Unfortunately, the old-fashioned
but inexpensive devices that follow these guide- floppy-arm device attached to a pole rising above
lines should give accurate measurements if the common office scale is used in many offices;
50 5 Disorders of Growth

because the plate at the top of the pole is rarely rounding error of one half-inch (1.25 cm) more
parallel to the floor, there is no way to straighten than the true measurement at the end of a 6-month
the patient’s back against a thin pole, and the child period can mean a fallacious 5 cm of increased
may slouch, and the measurements are useless. growth per year when there may be no growth
Weight measurements are performed on a digital due to a serious condition. Errors in the other
scale on a firm surface rather than carpet with direction can lead to an absence of 5-cm growth
coats and other heavy clothes removed. in a normal child, suggesting a disorder of growth
The child must be measured with the shoes when none exists.
off; if a child is measured in shoes one time and After the measurement is obtained, it must be
without them another time, a guaranteed 2–4-cm displayed graphically on the growth chart. An
variation in height is found per visit, and this abnormality of stature or growth rate is far more
all too often happens during casual evaluations. obvious on a graph than written as a number on
A great quantity of hair, braiding, or barrettes on the page; a decrease in growth rate in which a
the top of the head will likewise impair accuracy. child significantly “falls away from the curve” or
It is strongly recommended that all measure- “crosses percentiles of stature” becomes obvious
ments be made by using the metric system. The on the graph. The most commonly used charts of
tendency to round off numbers becomes problem- stature for age after 2 years of age (as well as
atic when an inch is the unit of measure; an inac- weight for age and BMI) from the National
curacy of an inch or a half inch is a more serious Center for Health Statistics are available at www.
error than a mistake of 1 or 0.5 cm; remember that cdc.gov; they may be downloaded or printed
a rounding error of one half-inch (1.25 cm) less (Fig. 5.4a, b). These reference charts are devel-
than the true measurement at the beginning and a oped from measurements of a population of US

Fig. 5.4 (a) Infant boy


growth chart developed by
the WHO as recommended
by the CDC for use in boys
under 2 years of age.
(b) Infant girl growth chart
developed by the WHO as
recommended by the CDC
for use in girls under 2
years of age. Accessed
from http://www.cdc.gov/
growthcharts/
Measurement of Growth 51

Fig. 5.4 (continued) Birth to 24 months: Girls


Length-for-age and Weight-for-age percentiles

98
95
90
75
50
25
10
5
2

98

95

90

75

50

25

10
5
2

Published by the Centers for Disease Control and Prevention, November 1, 2009
SOURCE: WHO Child Growth Standards (http://www.who.int/childgrowth/en)

children. The charts to be chosen show the 3rd to subtracting the birth date from the date of visit. If
97th percentile (not the 5th to 95th as they the month or the numerical day of the month of the
exclude too many normal children). The WHO birth date is greater than the month or the day of
produces standards of growth in optimal condi- the month of the visit, a conversion must be used
tions displayed on charts which are now the pre- to allow the subtraction to occur. One year is
ferred chart for children under 2 years of age also subtracted from the numerical year of the visit so
available at the CDC website (Fig. 5.4). Many that, e.g., 2014 is converted into 2013. One month
providers now use electronic medical records is subtracted from the numerical month of the visit
(EMRs) which readily display the percent of so that April which is represented by 4 is converted
height to a fraction which enhances diagnosis. to 3, and 12 months are added on to the result (3 in
The NCHS data tables which are available at this case) to represent 1 year that was taken off the
http://www.cdc.gov/nchs/data/series/sr_11/ numerical year. The value of the number of days in
sr11_252.pdf are used to compute standard devi- the month of the visit (e.g., 28 for February, 31 for
ation scores (SDS) for children falling below the July, 30 for November) is added to the numerical
3rd percentile. Specialty growth charts for Turner day of the month of the visit: February 8 is con-
syndrome, achondroplasia, and Down syndrome verted into 36. If the number of days in the result-
among others are available. ing age is between 0 and 15, the numerical value
When charting a child on a growth chart, the of the resulting age is left alone, but if the number
examiner must accurately determine the child’s of days and the resulting age are between 16 and
age at the visit date. EMR will usually calculate the end of the month, 1 month is added to the
the fractional age of the child thus eliminating the resulting age of the child. Thus a child born on
cumbersome process that follows. This involves 09/15/94 examined on 04/04/98 leads to a resulting
52 5 Disorders of Growth

age of 3 years, 6 months, and 19 days old. The of eunuchoid proportions of hypogonadism where
child described above is 3 years and 7 months old legs and arms are long). The arm span is mea-
by this calculation. Now the month of the child’s sured with the patient standing with the back to
age must be converted into fraction of the year to the wall with arms spread horizontally and is the
allow easy plotting; February to April is one-quar- distance from one outstretched middle fingertip to
ter of a year, April to July is one-half of a year, and the other. The arm-span measurement should be
August to October is three-quarters of a year, and close to the measurement of the height; on the
if the numerical month of birth of the child is average, normal prepubertal children have arm
November or December, represented by 11 or 12, span 1 cm less than their height, pubertal children
another year is added to round up to the next year have arm span equal to height, and adults have
of age. The child described above is 3 and one-half arm span 5 cm greater than height. If the arm span
years old which can be easily plotted on the growth is shorter than the height by more than the general
chart. (This entire process is described in the CDC guidelines above, a chondrodystrophy may limit
website at http://www.cdc.gov/nccdphp/dnpa/ long-bone growth, whereas if it is longer than the
growthcharts/resources/growthchart.pdf.) height, an abnormality of the growth of the spine
Growth velocity charts (Fig. 5.5) demonstrate may be present, perhaps due to spinal radiation
the rate of growth, are more sensitive to devia- therapy, among other possibilities. The lower seg-
tions from the normal growth velocity, and reveal ment is measured from the top of the symphysis
abnormalities before they become apparent on pubis to the floor, whereas the upper segment is
the standard height-for-age charts found in calculated by subtracting the lower segment from
Fig. 5.4 (see examples in Fig. 5.6). US growth the measured height of the child. The upper-to-
data were used to develop growth velocity charts lower segment ratio varies with age (Fig. 5.7). A
for US children; while 1985 charts used cross- decreased upper-to-lower segment ratio is found
sectional data, recently developed charts used in Klinefelter syndrome or other forms of hypo-
longitudinal data. gonadism due to longer legs, and an increased
Crossing percentiles on a growth chart can be ratio is found in hypothyroidism and chondrodys-
an ominous sign of disease but in infancy can be trophies due to poor growth of legs. Measurement
normal if the change is not excessive. Since the of occipitofrontal head circumference and deter-
length of healthy term babies clusters around mination of percent of SDS are part of the stan-
21 in. but adult heights are spread out wider, the dard examination using CDC charts as various
channel of growth followed on the growth curve central nervous system problems may lead to dis-
must change during the growing years, and this orders of growth (Fig. 5.8).
adjustment occurs before 2 years of age and usu- Skeletal development or bone age is sug-
ally before 1 year of age by a gentle deceleration gested to be more closely correlated with certain
or acceleration of growth rate. However, a flatten- developmental landmarks than with chronologic
ing growth curve or weight curve means signifi- age in conditions of delayed or advanced puberty.
cant organic disease. The difference between It is considered a reflection of the physiologic
these two patterns is not clear early in the course rather than the chronologic age of the child.
of the change in growth channels, and many However, careful study shows that the bone age
referrals to pediatric endocrinologists occur dur- does not always accurately reflect these goals, so
ing this period to rule out significant illness even the result should be interpreted with some suspi-
though the child is clinically well. cion. A bone-age determination does not provide
Determination of arm span and upper-to-lower a diagnosis but may support a condition under
segment ratio is useful in the evaluation of short consideration. The bone age is determined by a
stature (e.g., to indicate hypochondroplasia or radiograph of the left hand and wrist as com-
achondroplasia in which arms and legs are short) pared with the standards in the Greulich and Pyle
or of delay in puberty (e.g., to look for the long atlas in the USA, although Europeans may prefer
arms and decreased upper-to-lower segment ratio the Tanner–Whitehouse (TW2 or TW3) method.
Measurement of Growth 53

2 to 20 years: Boys NAME


Stature-for-age and Weight-for-age percentiles RECORD #

12 13 14 15 16 17 18 19 20
Mother’s Stature Father’s Stature cm in
Date Age Weight Stature BMI*
AGE (YEARS) 76
95
190
74
90
185 S
75
72
180 T
50 70 A
175 T
25 68 U
170 R
10 66
165 E
in cm 3 4 5 6 7 8 9 10 11 5
64
160 160
62 62
155 155
S 60 60
T 150 150
A 58
T 145
U 56
140 105 230
R
54
E 135 100 220
52
130 95 95 210
50
125 90 200
90
48 190
120 85
46 180
115 80
75
44 170
110 75
42 160
105 50 70
150 W
40
100 65 140 E
25
38 I
95 60 130 G
10
36 90 5 H
55 120
T
34 85 50 110
32 80 45 100
30
40 90
80 35 35 80
W 70 70
30 30
E 60 60
I 25 25
G 50 50
H 20 20
40 40
T
15 15
30 30
10 10
lb kg AGE (YEARS) kg lb
2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20
Published May 30, 2000 (modified 11/21/00).
SOURCE: Developed by the National Center for Health Statistics in collaboration with
the National Center for Chronic Disease Prevention and Health Promotion (2000).
http://www.cdc.gov/growthcharts

Fig. 5.5 (a) Height-for-age percentiles in boys from 2 to girls from 2 to 20 years (from CDC website, www.cdc.
20 years (from Centers for Disease Control website, www. gov/growthcharts)
cdc.gov/growthcharts). (b) Height-for-age percentiles in
54 5 Disorders of Growth

2 to 20 years: Girls NAME


Stature-for-age and Weight-for-age percentiles RECORD #

12 13 14 15 16 17 18 19 20
Mother’s Stature Father’s Stature cm in
Date Age Weight Stature BMI*
AGE (YEARS) 76
190
74
185 S
72
180 T
70 A
97 175 T
90
68 U
170 R
75 66
165 E
in cm 3 4 5 6 7 8 9 10 11 50
64
160 25 160
62 62
155 10 155
S 60 60
T 3
150 150
A 58
T 145
U 56
140 105 230
R
54
E 135 100 220
52
130 95 210
50
125 97 90 200
48 190
120 85
46 180
115 80
44 170
110 90 75
42 160
105 70
150 W
40 75
100 65 140 E
38 I
95 60 130 G
50
36 90 H
55 120
25 T
34 85 50 110
10
32 80 3 45 100
30
40 90
80 35 35 80
W 70 70
30 30
E 60 60
I 25 25
G 50 50
H 20 20
40 40
T
15 15
30 30
10 10
lb kg AGE (YEARS) kg lb
2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20
Published May 30, 2000 (modified 11/21/00).
SOURCE: Developed by the National Center for Health Statistics in collaboration with
the National Center for Chronic Disease Prevention and Health Promotion (2000).
http://www.cdc.gov/growthcharts

Fig. 5.5 (continued)


Measurement of Growth 55

Fig. 5.6 (a) Incremental growth charts for boys derived between measurements of height (g2 − g1) divided by
from data in Flemish children and generally applicable the size of the interval (t2 − t1). This formula is also
to the US population. Height velocity can be compared shown on the charts for yearly increments in height. It
with the percentiles. Increments in height are calcu- is important to plot the increments at an age which cor-
lated over periods of no less than 10.2 months (0.85 responds to the center of the interval, i.e., at the age
years) and no more than 13.8 months (1.15 years) in (t1 + t2)/2. Accessed at http://www.vub.ac.be/groeicur-
order to avoid the effect of seasonal variation in growth ven/files/2-20050604-EP2-20M.pdf and reproduced
and to reduce the effect of measurement error on the with premission. (b) Incremental growth charts for
estimation of yearly increment in stature. The yearly girls derived from data in Flemish children which
increment in stature is calculated as the difference should be generally applicable to the US population
56 5 Disorders of Growth

+3 +2 +1 0 −1 −2 −3
a In cm cm
60
59 150 150 −4
58
57 145 145 −5
56
55 140 140
54
53 135 135
52
51 130 130
50
49 125 125
48
47 120 120
46
45 115 115
44
Stature

110 110
43
42
41 105 105
40
100 100
39
38
95 95
37
36
90 90
35
34
85 85
33
32
80 80
31
30 Physical growth (NCHS)
75 75
29 Boys 2-18 y
28
70 70
27
In cm cm
2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18
Age (y)

Fig. 5.7 Demonstration of the sensitivity of the growth- growth chart of a child growing faster than normal early in
velocity charts compared to standard stature versus age life, who slows to a normal growth rate at 9 years and
charts. (a) The growth-velocity chart of the growth chart decreases thereafter, until ceasing to grow by age
with squares showing the more striking changes of growth ~13 years; this is found in untreated precocious puberty.
rate at various ages. The growth rate between two ages is (c) The growth-velocity chart of the boy with the growth
plotted as a horizontal bar, with the height of the bar dem- chart with circles. The decrease in growth rate is more
onstrating the annualized growth rate and the lateral limits striking earlier than is apparent on the statural growth
spanning the ages under consideration. (b) A growth chart chart. The growth rate between two ages is plotted as a
of two boys. The circles represent a boy who has a pro- horizontal bar, with the height of the bar demonstrating
found decrease in growth rate at age 3–4 years. At age 6 the annualized growth rate and the lateral limits spanning
years, some intervention occurs (red arrow), and growth the ages under consideration. Redrawn from Styne DM:
catches up at greater-than-average growth velocity. This Growth. In: Greenspan FS and Strewler FJ, eds.
could be the institution of thyroxine in hypothyroidism, Greenspan’s Basic and Clinical Endocrinology, 9th edi-
growth hormone (GH) in GH deficiency, or removal of a tion. Appleton and Lange, Stamford, Connecticut. 2011
glucocorticoid-secreting tumor. The squares represents a with permission
Measurement of Growth 57

Stature
boys
b 6

4
Increment (cm/6 months)

97
2

90
1
75
50
0 25
10

3
–1
2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18
Age (y)

Stature
boys
c 6

4
Increment (cm/6 months)

97

90

1 75
50
25
0
10

3
–1
2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18
Age (y)

Fig. 5.7 (continued)


58 5 Disorders of Growth

1.20 increase accuracy of the reading. Of course, the


closer a child’s age is to adulthood, the more
accurate the prediction for adult height. A bone
1.10
age is not a perfect test, but the information gar-
nered is nonetheless of use in evaluating a child.
Bone age will be delayed in malnutrition as well
+2 SD as in some endocrine diseases. Bone age will be
1.00 advanced in many cases of obesity reportedly more
+1 SD prevalent in MC4R mutations.
US:LS

Mean Long-bone X-rays may demonstrate growth


arrest (or Harris or Park lines) which are dense
0.90 –1 SD
metaphyseal, trabecular lines thought to occur
–2 SD after normal growth resumes, after nutritional
deprivation, immobilization, and illness includ-
ing infectious disease in growing children.
0.80

Endocrine Factors in Postnatal


Growth
4 6 8 10 12 14 16 18
Age (y)
Growth Hormone
Fig. 5.8 Upper-to-lower segment ratio (redrawn from
Heritable Disorders of Connective Tissue St. Louis: C. V. Growth hormone (GH) is a single-chain 191-
Mosby Company, 1972 , with permission)
amino-acid peptide hormone with two disulfide
bonds produced in the anterior pituitary gland. It
The delay or advancement in bone age is resembles prolactin, chorionic somatomam-
expressed in standard deviations from the mean motropin, or placental lactogen in structure.
reading for chronologic age which is listed in the While primate growth hormone will exert effects
Greulich and Pyle atlas. Standard deviations on other mammals, e.g., cows, bovine growth
vary with age; 1 standard deviation may be only hormone will not exert metabolic effects in
2 months during the first year up to a standard human beings, and thus natural sequence human
deviation of 1 year at 15 years (this means that a growth hormone is necessary for treatment in
normal 15-year-old boy may have a bone age children. Production and secretion of GH are
between 13 and 17 years!). Increased accuracy stimulated by growth hormone-releasing factor
in the technique is accomplished by radiologists or GHRH (or GRF in some texts) which is pro-
with the most practice; unfortunately, most gen- duced in the median eminence of the hypothala-
eral radiologists who are not associated with a mus (Fig. 5.9a). GH deficiency may be caused by
pediatric center do not read many bone ages, and GHRH deficiency or GH deficiency (Fig. 5.9b).
their readings may not be accurate. With a bone GH secretion is suppressed by somatostatin
age over 6 years and an accurate determination (growth hormone-releasing inhibitory factor or
of stature, adult height can be predicted by the SRIF or SS) which has far-ranging effects in the
Bayley–Pinneau tables found at the end of the body including inhibiting the secretion of TSH,
Greulich and Pyle atlas. The Roche–Wainer– insulin, and glucagon; impairment of gut motil-
Thissen (RWT) method allows prediction of ity; and reduction of intestinal absorption of
eventual adult height in younger children. Other nutrients. The balance between GHRH and SRIF
methods of height prediction are in use in leads to the pulsatile release of GH in normal
Europe, and computerized methods of bone-age individuals. GH secretion is inhibited by insulin-
evaluation are used in Europe in an attempt to like growth factor I (IGF-1) through negative
Endocrine Factors in Postnatal Growth 59

a Third ventricle Hypothalamus

Neuroendocrine
cell nuclei

GHRH secreting cells

Superior
hypophyseal Stalk
artery

Long portal
vessels Inferior
hypophyseal
artery
Somatotrophs

Anterior Posterior
pituitary pituitary

Short portal
Hormone vessel
secretion

GH

IGF-1

Fig. 5.9 (a) Normal growth hormone physiology in which absence of growth hormone, no IGF-1 is produced and both
growth hormone-releasing hormone (GHRH) from the growth hormone and IGF-1 levels are low, leading to
hypothalamus reaches the pituitary gland through the hypo- growth hormone deficiency. (c) Growth hormone resis-
physiotropic portal system to cause growth hormone secre- tance in which growth hormone receptors are either absent
tion. Growth hormone interacts with its receptors on the or defective. Thus no IGF-1 can be produced and there is
liver and other organs to produce insulin-like growth factor no negative feedback inhibition on the pituitary gland or
I (IGF-1) which exerts influences on cells throughout the hypothalamus. GHRH and growth hormone rise in an
body and provides feedback inhibition on the pituitary unsuccessful attempt to stimulate IGF-1 production, lead-
gland and hypothalamus to suppress GHRH and growth ing to the elevated growth hormone levels and depressed
hormone secretion until a normal equilibrium is reached. IGF-1 levels characteristic of growth hormone resistance.
(b) Growth hormone deficiency due to the absence of This condition is also called Laron dwarfism or primary
growth hormone-producing cells, somatotrophs. The same IGF-1 deficiency. Similar changes occur in starvation in
physiology applies if GHRH cells are defective. In the which GH receptors are decreased

feedback inhibition. GH itself inhibits GH secre- Growth hormone exerts direct effects but also
tion, providing another level of control of GH produces effects through the production of IGF-1
secretion. In states of IGF-1 deficiency such as in an indirect manner. Acute effects of GH admin-
GH receptor mutations, GH levels rise (Fig. 5.9c). istration are insulin-like, as GH increases amino
60 5 Disorders of Growth

b Third ventricle Hypothalamus

Neuroendocrine
cell nuclei

GHRH secreting cells

Superior
hypophyseal Stalk
artery

Long portal
vessels Inferior
hypophyseal
artery
Somatotrophs

Anterior Posterior
pituitary pituitary

Short portal
Hormone vessel
secretion

GH

IGF-1

Fig. 5.9 (continued)

acid uptake and incorporation into protein in the GH secretion is high in the 48 h after birth,
liver and muscle, stimulates glucose uptake and decreases in the child, and rises during puberty
glucose utilization, and antagonizes the lipolytic as, with rising IGF-1, it mediates the pubertal
effects of catecholamines. Chronic effects of growth spurt. Sex steroids affect growth hormone
GH are diabetogenic or anti-insulin and include secretion, and it is estradiol that increases growth
increased mobilization of FFA from adipose tis- hormone secretion during puberty in both boys
sue due to increased triglyceride lipolysis, and girls. GH decreases with aging in normal
increased sensitivity to lipolytic effects of adults. GH rises with stress, slow-wave sleep,
catecholamines, as well as inhibition of glucose exercise, hypoglycemia, and fasting and decreases
uptake and utilization. GH indirectly stimulates with obesity, sugar ingestion, rising free fatty
insulin secretion via the increase in serum glucose acids, exogenous growth hormone administra-
concentrations. tion, and REM sleep. Estradiol stimulates GH
Endocrine Factors in Postnatal Growth 61

c Third ventricle Hypothalamus

Neuroendocrine
cell nuclei

GHRH secreting cells

Superior
hypophyseal Stalk
artery

Long portal
vessels Inferior
hypophyseal
artery
Somatotrophs

Anterior Posterior
pituitary pituitary

Short portal
Hormone vessel
secretion

GH

IGF-1

Fig. 5.9 (continued)

secretion in moderate doses but suppresses GH (GHS-R1a). Galanin is another hypothalamic


secretion at high levels. Estradiol has similar peptide that stimulates growth hormone release
effects on IGF-1 levels. without affecting other pituitary hormones.
Synthetic six-amino-acid peptides (hexapep- GH exerts its effects through the cell mem-
tides) are also known to stimulate GH release and brane GH receptor (see Fig. 1.3). There is a com-
have been used as GH secretagogues in testing. plex chain of events that occur once GH is
The study of these factors led to the discovery of available to bind to GH receptors, and all can go
the endogenous growth hormone secretagogue, awry in disorders. The growth hormone (GHR)
ghrelin, which is produced in the gastrointestinal receptor, once bound to a single GH molecule,
tract; ghrelin is considered an orexigenic or appe- dimerizes with another unoccupied receptor,
tite-stimulating hormone; the natural receptor for leading to a complex of one GH molecule and
ghrelin is the GH secretagogue receptor type 1a two GHR molecules in a dimeric complex.
62 5 Disorders of Growth

This complex causes activation of the Janus when elevated levels of GH will be diagnostic;
kinase-signal transducer and activator of tran- only sequential measurements of GH through a
scription (Jak-STAT) pathway. Recruitment of 12- or 24-h period (rarely performed because of
Janus kinase 2 (JAK2) stimulates tyrosine kinase. expense) or measurements after the administra-
The phosphorylated receptor provides a site for tion of a GH secretagogue are of interest in the
STAT to attach and become phosphorylated diagnosis of GH deficiency. However, even
itself. The phosphorylated STAT molecule then secretory tests are suspect, as some children
dissociates from the receptor site to move to the with normal GH secretion on testing can benefit
nucleus where it binds to target genes and regu- from GH treatment, and others with low values
lates transcription to produce proteins. on such a test grow normally and need no GH.
The amino acid sequence of the extra- In GH deficiency, bone-age development is
membrane portion of the GH receptor is cleaved delayed, whereas the upper-to-lower segment ratio
off to give rise to the GH-binding protein (GHBP) is normal for bone age. This is in contrast to hypo-
found in the circulation. About ½ of circulating thyroidism, in which bone age is severely delayed,
GH is noncovalently bound to GHBP. The abun- but the upper-to-lower segment ratio is increased
dance of the membrane receptor for GH is due to lack of appropriate growth of the lower
reflected by the value of circulating GHBP. In limbs. Patients with untreated GH deficiency may
classic GH resistance (e.g., Laron dwarfism) in have delay in the onset of puberty until the absent
which there are decreased or no cell membrane GH is replaced with treatment, and puberty pro-
growth hormone receptors, the serum GHBP val- gresses. It is thus difficult to determine which
ues are low, whereas serum GH is high. In other patient with delayed puberty and untreated GH
forms of GH resistance, the number of receptors deficiency has only GH deficiency and which has
and the measurement of GHBP are normal, but gonadotropin deficiency in addition.
the function of the receptor is defective (Fig. 5.9c).
Growth hormone-binding protein is decreased in
starvation, indicating that a decrease in growth Insulin-Like Growth Factor or IGF
hormone receptors occurs in a situation where
growth hormone is elevated so that GH can exert GH has some direct metabolic effects (e.g., diabe-
no effect on growth. On the contrary, increased togenic insulin-resistant effects, lipolysis, cell dif-
body mass index and obesity increase growth ferentiation) and exerts direct growth-promoting
hormone receptors as reflected in increased effects on the growth plate, but much of the
growth hormone-binding protein levels and sub- growth-promoting action of GH is indirectly medi-
sequent normal or even high normal IGF-1 val- ated by insulin-like growth factor 1. Growth fac-
ues even though growth hormone is low. tors can be secreted to affect other cells with
Every step in GH action is subject to a endocrine activity and affect neighboring cells
genetic error that will lead to short stature. For with paracrine activity or the cell of origin with
example, mutation of STAT 5b leads to short autocrine activity or intracrine within a cell.
stature that is GH resistant and is associated Insulin-like growth factors (IGFs, previously
with immune deficiency (#245590 GROWTH called somatomedins) are produced in tissue
HORMONE INSENSITIVITY WITH throughout the body, but most IGF-1 is detected in
IMMUNODEFICIENCY). Serum GH concen- the serum derives from the liver. IGF-1 has mito-
trations are low throughout most of the day in genic effects in most dividing cells in the body and
normal individuals but increase at intervals, stimulates sulfate uptake (and it was originally
and these peaks differentiate a GH-sufficient known as sulfation factor), DNA synthesis, RNA
patient from a GH-deficient subject in most synthesis, and protein synthesis and exerts anti-
cases. Thus, random GH measurements do not apoptotic effects. IGF-1 values are most closely
reveal abnormalities of secretion unless GH associated with the growth of children. Plasma val-
resistance or pituitary gigantism is suspected ues of IGF-1 decrease in states of GH deficiency
Endocrine Factors in Postnatal Growth 63

and increase in GH excess. IGF-1 concentrations IGFBPs characteristically decrease the activity of
are low in the neonate and increase slowly through IGF-1 by holding it in the reservoir of inactive
childhood until a peak is reached during the puber- complex but in some cases, others can enhance the
tal period with an earlier peak in girls than boys, effects of IGF-1. For example, binding proteins
reflecting the differential timing of the pubertal can also allow IGF-1 to transit cell membranes in
growth spurt in the sexes. Normal values of IGF-1 some cases. IGFBP-3 is GH dependent, and low
at puberty are in the range characteristic of acro- serum values in association with low serum values
megalic adults. The major problems with the inter- of IGF-1 make the diagnosis of GH deficiency
pretation of IGF-1 concentrations as a reflection of more likely than a decrease in serum IGF-1 by
GH secretion are the following: (a) the values in itself, especially in children before 3 years of age
the first 3 years of postnatal life are close to those when IGF-1 values are normally low. IGFBP3 is
found in the hypopituitary state; (b) the values in also responsive to IGF-1 so that values of IGFBP3
constitutional delay in growth are appropriate for rise in conditions of GH and/or IGF-1 excess.
bone age rather than chronologic age and are often About 80–85 % of serum IGF-1 and IGF-2 are
reported as abnormally low if correlated with found in a ternary complex of 150 kDa composed
chronological age alone while, if corrected for of one molecule each of IGF-1, IGFBP3, and a
bone age, the values would be reported as normal; protein that is found only in serum, the acid-labile
and (c) in states of poor nutrition, a condition that subunit (ALS). The tertiary compounds of IGF 1,
is extremely common that by itself can cause poor IGFBP3, and ALS reflect growth hormone secre-
growth, IGF-1 values are as low as those in a tory levels.
GH-deficient subject. Of further importance is the On the contrary, IGF-binding protein 1 is insu-
variation between measured IGF-1 concentrations lin dependent but not GH dependent. In states of
in different laboratories and the lack of the stan- hyperinsulinism, IGFBP 1 is suppressed.
dardized assay. Thus the use of serum IGF-1 con- IGF-1 attaches to a specific IGF-1 receptor which
centrations must be tempered with caution in the is structurally similar to the insulin receptor (Fig.
diagnosis of growth deficiency, and strict attention 1.3). Cross-reaction of insulin with IGF-1 receptors
must be paid to nutritional status and physiologic is possible when insulin levels are elevated as in an
state of development. Protein calorie malnutrition infant of a diabetic mother. Alternatively, the admin-
decreases plasma IGF-1 values due to decreased istration of IGF-1 may cause low blood sugar due to
binding of GH to the GHR. Further IGF-1 mea- interaction with insulin receptor.
sured in the serum is only a reflection of local Some suggest the abandonment of secretory
activity within tissue of origin the growth factor. tests of GH secretory ability and argue that reli-
IGF-1 production is found in certain cancer ance on serum IGF-1 and IGFBP-3 is sufficient for
cells. Serum levels of IGF-1 may be related to diagnosis of GH deficiency. In California, some
some malignancies. insurance companies have adhered to this view,
Insulin-like growth factors I and II are bound to and the state funding agency, California Children’s
one of a family of more than six IGF-binding pro- Services, at present has ceased requiring GH stim-
teins, each identified with a different number such ulatory testing to establish GH deficiency in most
as IGFBP1, IGFBP2, and IGFBP3. The structure cases. However, not all insurance companies or
of the different IGF-binding proteins are relatively state agencies agree to pay for GH treatment with-
similar but quite different than growth hormone- out a GH secretory test, so such stimulatory testing
binding protein or steroid-binding proteins. procedures cannot be abandoned.
Binding proteins keep IGF-1 levels relatively con- IGF-1 is important in late fetal growth as an
stant throughout 24 h in contrast to the episodic individual with an IGF-1 loss of function receptor
secretion characteristic of growth hormone. IGF- mutation suffered from severe IUGR. As growth
binding protein 3 measurements can be indicative hormone receptors are low in the fetus compared
of GH status. IGF-binding protein can modify the to postnatal life, it is insulin that regulates IGF-1
biological effects of insulin-like growth factor. in the fetus rather than growth hormone.
64 5 Disorders of Growth

IGF-2 is present in high concentrations after and hypothyroidism may be confused in this way
birth but does not have the relationship to with growth hormone deficiency. IGF-1 levels
growth found with IGF-1. Concentrations of are also low in hypothyroidism.
IGF-2 are decreased in GH deficiency but not Sex steroids advance growth, skeletal age, and
increased in GH excess. Although plasma pubertal development in the postnatal state.
IGF-2 concentrations alone may not be reliable Gonadal steroids are essential for the pubertal
in the diagnosis of GH deficiency, the finding of growth spurt and are responsible for approximately
both low values of IGF-1 and IGF-2 has been half of the growth achieved during puberty. If
stated to be reliable in pointing to the diagnosis excessive sex steroid concentrations are maintained
of growth hormone deficiency. In general, how- for a long period (e.g., virilizing congenital adrenal
ever, IGF-2 measurements are not used clini- hyperplasia), the epiphyses will fuse prematurely,
cally in the diagnosis of disorders of growth and the previously tall child will become a short
although IGF-2 does play a role in neoplasia adult; epiphyseal fusion is owing to the action of
and in the evaluation of carcinoma as, e.g., lev- estrogen, which may be secreted directly or result
els may be increased in patients with osteosar- from the aromatization of testosterone. Absence of
coma. The insulin-like growth factor 2 receptor gonadal steroids will delay skeletal maturation and
(IGF2R encoded by the IGF2R gene) is quite decrease the pubertal growth spurt, but hypogonad-
different in structure and function from the ism can also allow prolonged growth, leading to
IGF-1 receptor and is also known as the cation- taller-than-predicted adult stature.
independent mannose-6-phosphate receptor Glucocorticoids significantly suppress growth
(CI-MPR) indicating its other functions. when present in excess; thus, Cushing syndrome
Thyroid hormone is essential for postnatal due to endogenous (e.g., an adrenal adenoma) or
growth but has little effect on fetal longitudinal exogenous (e.g., given for rheumatoid arthritis or
growth, as congenital hypothyroidism is associ- renal disease) glucocorticoids can cause the ces-
ated with normal or even slightly greater than sation of growth and result in short stature. Inhaled
normal birth length. Thyroid hormone is also glucocorticoids for asthma therapy may have
necessary for the normal secretion of GH, as measurable but smaller effects on growth than do
hypothyroid patients may not respond to GH oral glucocorticoids. Even dermal application of
stimulation tests with GH secretion, thereby glucocorticoids in excess can affect growth, espe-
confusing the diagnosis of hypothyroidism with cially if the skin barrier is breached. If glucocorti-
GH deficiency. Hypothyroid patients who also coids for treatment of a chronic disease can be
have GH deficiency cannot respond to GH ther- minimized by the use of decreased dosage, alter-
apy unless they are rendered euthyroid. In the nate-day treatment, or even alternative therapy,
absence of adequate thyroid hormone, a patient growth will usually benefit. Decreased glucocorti-
has reduced lower limb growth, leading to a coids will not affect growth as long as the patient
delayed or higher upper-to-lower segment ratio. is not disabled or anorexic from the condition.
Bone-age development is also retarded in hypo- Insulin in excess will increase growth rate in
thyroidism, and when the epiphyses do appear, the fetus (e.g., infant of a diabetic mother) and
they are often irregular and abnormal, with mul- after birth (e.g., patients with insulinomas or islet
tiple epiphyses (epiphyseal dysgenesis). Usually cell hypertrophy) due to interaction of insulin
puberty is delayed if thyroid hormone is with IGF-1 receptors. Insulin deficiency will
decreased, but with profound primary hypothy- decrease growth if replacement is inadequate
roidism, pubertal development may occur early, because of the many serious effects of diabetes.
particularly in girls, apparently due to cross-reac- Further, in the absence of insulin receptors, as in
tion of elevated TSH values with gonadotropin Donohue’s “leprechaun” syndrome (#246200
receptors; this is known as the Van Wyk-Grumbach ICD+DONOHUE SYNDROME at 19p13-2), an
syndrome (see Chaps. 7 and 8). Hypothyroidism autosomal recessive condition, fetal and postna-
inhibits the ability to secrete growth hormone, tal growth will be poor.
Endocrine Factors in Postnatal Growth 65

Fig. 5.10 Calculation of target height for children by using increased by 5 in. on a boyl’s chart (upper thin red line).
the stature-for-age percentiles for boys aged 2–20 years The midparental height (MPH) (thick red line) is the aver-
growth chart (from CDC website, www.cdc.gov/ growth- age of the father’s height and the corrected mother’s height.
charts). The father’s height (in yellow) is plotted on the right The limit of 2 standard deviations (SD) is approximately 4
directly, while the mother’s height (lower thin red) must be in. above and below the MPH indicated by the black lines

Genetic factors are of significant importance adjusted height appropriately on a daughter’s


in growth as it is estimated that about 40–50 % of chart (as shown in Fig. 5.10) or 5 in. is added to
stature is inherited. The correlation between a mother’s height to allow plotting her adjusted
midparental height and children’s height can be height on a son’s chart (plotted at 18–20 years,
used to adjust the position of a child on a growth an age when growth normally ceases and is the
chart to better reflect the genetic milieu of the adult height). Then the father’s height and
family. The average difference in adult stature of adjusted mother’s height are averaged, and the
about 13 cm which is usually rounded out to 5 in. midparental height that results (the adjusted
between men and women in the USA is used for midparental height) is plotted on the far right of
the process; thus 5 in. is subtracted from a the son’s growth chart (e.g., at 18 years, or adult
father’s height for a girl to allow plotting his stature) or the mother’s height and the adjusted
66 5 Disorders of Growth

father’s height are averaged, and the resulting stimulatory testing as they will not normally raise
midparental height plotted on the far right of the their growth hormone values; the test results will
daughter’s growth chart (Fig. 5.10). The limits of likely revert to normal by moving the child out of
2 standard deviations (in the USA, a standard the home and into a more welcoming environ-
deviation is about 2 in. for adult height, so 2 SD ment where growth may again resume.
is 4 in.) or 4 in. are noted above and below the
midparental height. This then is the target height
of the child. One can conceptually adjust the Epidermal Growth Factor
growth chart for the family by interpreting the
midparental height as the 50th percentile for this Epidermal growth factor (EGF) binds to its own
particular family and interpreting the height of EGF receptor (EGFR), a member of the ErbB fam-
the child by the resulting new percentiles on the ily of receptors that have tyrosine kinase activity,
conceptual chart. Thus, a boy who plots at the leading to dimerization of the receptors causing
3rd percentile for the USA who has short parents stimulation of cell growth, proliferation, and
might plot well within the target height (or on the differentiation. In the normal state epidermal growth
conceptually reinterpreted chart at the 10th per- factor is a mitogen for ectodermal tissue but in neo-
centile for the family; Fig. 5.10). plastic conditions, excess epidermal growth factor
Nutrition is one of the most essential factors for may be produced.
growth and reproductive development. In the eval-
uation of children immigrating to the USA from
developing countries or from countries torn by war Erythropoietin
or famine, it may be inappropriate to consider the
parents’ heights in the evaluation of the child; the Erythropoietin is the growth factor produced in the
parents may have been subject to malnutrition dur- interstitial fibroblasts in the kidney which stimu-
ing their growing years and may be inappropri- lates red blood cell production. Erythropoietin val-
ately short, whereas the child may benefit from ues rise in anemia and hypoxemia. Erythropoietin
better nutrition in their new home and have the is also stimulated by thyroid hormone, adrenal
opportunity to be taller than the parents. Decrease cortical steroids, androgens, and growth hormone.
in nutrients from excessive dieting or anorexia ner-
vosa as well as chronic disease can exert the same
effect as malnutrition due to economic causes on Oncogenes
decreasing growth. Thus, socioeconomic and psy-
chosocial factors are important considerations in Oncogenes are any genes that encode a protein
the interpretation of growth rate. Of course it is able to transform cells to induce cancer. Oncogenes
important to ensure that the parents reporting their are able to stop cell from undergoing programmed
heights are indeed the biological parents, informa- cell death, apoptosis, often in concert with envi-
tion that might not be volunteered automatically. ronmental factors or viruses. Most oncogenes
Chronic disease may decrease growth apart originate from normal genes, proto-oncogenes,
from effects on decreasing nutritional intake. For which encode for positive-acting growth factors
example, the decrease in stature in children with and their receptors, signal-transduction proteins,
juvenile rheumatoid arthritis and celiac disease is transcription factors, and cell-cycle control pro-
not explained solely on nutritional grounds. teins. When proto-oncogenes undergo activation
Psychological problems can affect growth or mutation, they produce the tumor-causing onco-
either from a nutritional standpoint or through an genes. An oncogene may cause the growth factor-
endocrine effect. Psychosocial dwarfism is a producing cell to secrete growth factors in
temporary condition of hypopituitarism precipi- increased amounts or to make cells that are not
tated by abnormal parent-child interaction which supposed to produce growth factors to secrete
is not thought to be due to malnutrition. Affected them. Oncogenes are similar to genes that nor-
children will test positive for GH deficiency on mally function in fetal development: for example,
Abnormalities of Growth 67

the WT 1 or Wilms tumor gene is implicated in purpose of the definition: the Growth Hormone
numerous other types of tumors as well. However, Research Society uses <3 SD below the mean
the WT 1 gene is essential for the development of or the <0.1 percentile for idiopathic short stat-
many organs and tissues in the fetus. ure (ISS), the Pediatric Endocrine Society
uses <2 SD below the mean for age and gen-
der or 2.3 %, and the FDA uses −2.25 standard
Dental Development deviations for consideration of growth hor-
mone therapy for ISS. For the purposes of
Orthodontic treatment is predicated upon the this chapter we will adhere to the −2 standard
development of teeth, but a remarkable and com- deviation definition which is the 2.3 percentile
prehensive theory that controls the development and which comes close to the 3rd percentile
of teeth is not available. It is useful however to line on a CDC growth chart.
predict the likelihood of future dental develop- 2. Height velocity <2 SD below mean for bone
ment in delayed puberty, growth hormone defi- age.
ciency and short stature. Certain milestones in 3. Height corrected for midparental height of
dental development are reflected by the age of <−2 SD or <2.3 % (the Growth Hormone
tooth eruption but this is a single event in the his- Research Society uses <1.5 SD).
tory of tooth, while calcification of the tooth 4. Signs indicative of an intracranial lesion.
might be considered a more reliable indicator. 5. Signs of multiple pituitary hormone deficiency.
Various methods of estimating dental maturity 6. Neonatal symptoms and signs of growth hor-
using either eruption or calcification are avail- mone deficiency.
able, and some data is available on the effect of 7. History of central nervous system radiation.
tooth development in endocrine disorders. It is
generally agreed that the first permanent teeth The more of the criteria met, the more likely
erupt between an average age of 6 and 7 years an abnormality.
and that the first teeth are the first molar and lat-
eral/central incisors which emanate from differ- Nonendocrine Causes of Short Stature
ent tooth groups. Either eruption may precede the
other and the eruptions are uncoordinated. Constitutional Delay in Growth
Growth hormone-deficient and growth hormone- Constitutional delay in growth and adolescence
resistant children have delayed dental eruption, (CDP or constitutional delay) is best considered a
but GH treatment does not usually decrease the variation of normal development (Table 5.1). After
degree of delay in dental development. Severe a normal birth length and weight, growth rate sub-
hypothyroidism is associated with severe delay in tly decreases during the first 2 years so that stature
dental development. becomes shorter than average for age 2–3 years,
but the child then follows the channel of growth on
the chart just reached (i.e., growth rate then nor-
Abnormalities of Growth malizes (Fig. 5.11)). In addition, bone age is
delayed at least 2 SDs by the definition of constitu-
Short Stature tional delay. Thus the condition is a variation in the
normal tempo of development rather than a dis-
The criteria for awakening concern over a disor- ease. The child is usually thin, often male, but with
der of growth depend upon the defining group the obesity epidemic, a subgroup of obese consti-
(Pediatric Endocrine Society, FDA, Growth tutional delay has emerged, so weight is no longer
Hormone Research Society), but for this discus- an accurate indicator. The growth rate is appropri-
sion, we suggest evaluation if: ate for the skeletal development but not for the
chronologic age. Thus the growth rate must be
1. The definition of short stature varies by the interpreted in terms of bone age rather than chron-
organization making the definition and the ologic age. If the patient has genetic short stature
68 5 Disorders of Growth

Table 5.1 Etiologies of short stature Table 5.1 (continued)


Variations of normal Noonan syndrome (pseudo-Turner syndrome)
Constitutional delay in growth and puberty Autosomal trisomy 13, 18, 21
Genetic short stature with short familial heights Prader-Willi syndrome
Endocrine disorders Bardet-Biedl syndromes
GH deficiency Autosomal abnormalities
Congenital Dysmorphic syndromes (Russell-Silver, Cornelia de
Isolated GH deficiency Lange)
With other pituitary hormone deficiencies Pseudohypoparathyroidism/Albright’s hereditary
With midline defects osteodystrophy
Pituitary agenesis Chronic disease
With gene deficiency Cardiac disorders
Acquired Left-to-right shunt
Hypothalamic/pituitary tumors Congestive heart failure
Histiocytosis X (Langerhans cell histiocytosis) Pulmonary disorders
CNS infections and granulomas Cystic fibrosis
Head trauma (birth and later) Asthma
Hypothalamic/pituitary radiation GI disorders
CNS vascular accidents Malabsorption
CNS surgery Celiac disease
Hydrocephalus Disorders of swallowing
Empty sella syndrome Inflammatory bowel disease
Autoimmune hypophysitis Hepatic disorders
Functional GH deficiency Hematologic disorders
Psychosocial dwarfism Sickle cell anemia
GH resistance or primary IGF deficiency Thalassemia
Various forms of GH resistance Renal disorders
Laron dwarfism (increased GH and decreased IGF-1) Renal tubular acidosis
Pygmies (normal GH and IGF-2 but decreased IGF-1) Chronic uremia
IGF 1 resistance Immunologic disorders
Hypothyroidism Connective tissue disease
Glucocorticoid excess Juvenile rheumatoid arthritis
Endogenous Chronic infection
Exogenous AIDS
Diabetes mellitus under poor control Hereditary fructose intolerance
Diabetes insipidus (untreated) Malnutrition
Hypophosphatemic vitamin D-resistant rickets Fad diets and anorexia nervosa
Virilizing congenital adrenal hyperplasia (tall child, Kwashiorkor, marasmus
short adult) P-450c21, P-450c11 deficiencies Iron deficiency
Skeletal dysplasias Zinc deficiency
Osteogenesis imperfecta Anorexia due to chemotherapy for neoplasms
Osteochondroplasias Amphetamine treatment for hyperactivity with
Lysosomal storage diseases decreased caloric intake
Mucopolysaccharidoses Modified from Styne DM. Growth disorder. In: Fitzgerald
Mucolipidoses PA, ed. Handbook of clinical endocrinology. Norwalk,
CT: Appleton & Lange, 1986: 73–99, with permission
Syndromes of short stature AIDS acquired immunodeficiency syndrome, CNS central
Turner syndrome (syndrome of gonadal dysgenesis) nervous system, GI gastrointestinal, IGF insulin-like
SHOX deficiency growth factor, GH growth hormone
cm In In
200
78 78
195
76 76
190
Stature-for-age percentiles: 97th
74 Boys, 2 to 20 years 95th
90th
74
185
72 72
75th
180
70 70
50th
175
68 68
25th
170
66 10th 66
165 5th
64 3rd 64
160
62 62
155
60 60
150
58 58
145
56 56
140
54 54
135
52 52
130
50 50
125
48 48
120
46 46
115
44 44
110
42 42
105
40 40
100
38 38
95
36 36
90

34 34
85

80 32 32

30 30
75
cm In In
2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20
Age (years)
Published May 30, 2000.
SOURCE: Developed by the National Center for Health Statistics in colaboration with
the National Center for Chronic Disease Prevention and Health Promotion (2000).

Fig. 5.11 Growth chart demonstrating the various patterns his adult height may not reach his genetic potential as indi-
of growth. A: Blue line. A boy with genetic short stature who cated by parental heights. C: Red line. Growth of a boy with
parallels the normal curve below the 3rd percentile and who a serious disease, which could be a brain tumor that interferes
has no delay in bone age. He reaches an adult height at a with growth hormone secretion, glucocorticoid-induced
percentile similar to the percentile at which he grew during growth failure, or a host of other chronic diseases. His growth
childhood. B: Green line. A boy with constitutional delay in decreases from about the 50th percentile remarkably at about
growth, who parallels the normal growth curve but is below age 10 years, which is the age at onset of his disorder. He
it in a manner similar to a boy with genetic short stature but should have been diagnosed with a problem soon after the
has a delayed bone age, and so has a late pubertal growth change in growth velocity based upon this plot but the line is
spurt and continues to grow after the average boys have extended as if he was not diagnosed and treated, for emphasis
stopped. He will reach an adult height in the normal range (drawn on chart redrawn from CDC website, www.cdc.gov/
above the percentile he followed during childhood; however growthcharts)
70 5 Disorders of Growth

in addition to constitutional delay in growth, the delay in growth in their patient populations confus-
degree of short stature may be so obvious that the ing the conclusions. The FDA defines children with
patient will seek advice sooner; if constitutional ISS as being −2.25 SD below the mean or below
delay occurs in a child without genetic short stat- the 1.2 % which is well below the 3rd percentile
ure, the degree of short stature may not stimulate a shown on standard growth charts. Further, if an
medical evaluation until a delay in puberty is otherwise normal child has a predicted height less
noted. Often one parent or a sibling has a history of than 63 in. (160 cm) for males and less than 59 in.
constitutional delay (e.g., mother did not begin (149 cm) for girls, they meet the FDA approved
menstruating until 15–16 years or father continued criteria for growth hormone treatment, but not all
to grow or started to shave well after high school). insurance companies will pay for such treatment.
Patients with constitutional delay will enter A review of randomized controlled studies and
puberty at a later age than their peers (by defini- other studies indicates a mean adult height gain of
tion, older than 13 years in girls and 14 years in 5.5 cm with treatment if started by 10 years of age.
boys) but at an age appropriate for their bone age.
Although considerable variation exists, as a rule Small for Gestational Age
of thumb, boys will enter puberty when their Small for gestational age (SGA) infants are born
bone age is 12 years and girls when their bone with birth weights below the tenth percentile for
age is 11 years. The predicted height of children their gestational age; for a term baby, this translates
with significant delay in bone age calculated by into an approximate weight less than 2,500 g
the use of the Bayley–Pinneau tables in the (although there are other weight definitions pro-
Greulich and Pyle atlas may overestimate adult posed in various publications). Standards are avail-
height; thus their adult height, although within able (e.g., Fig. 5.1a) for weight according to
the normal range, might be lower than expected. gestational age to interpret the birth weight of pre-
mature infants. Intrauterine growth-retarded or
Genetic Short Stature growth-restricted (IUGR) babies have poor intra-
Genetic short stature refers to the child of shorter- uterine growth monitored by ultrasound and usually
than-average parents. These children are expected are born SGA. The terminology is often confused in
to reach an adult height below average, as their various texts, but we adhere to the definition of
short stature is not accompanied by a delay in bone SGA based on birth weight compared with gesta-
age. Adjustment for midparental height is of impor- tional age and IUGR based on fetal growth rate.
tance in evaluating such children (Fig. 5.10). Symmetrical SGA indicates that a decreased head
It bears mentioning that the heights of the biologi- circumference accompanies the decreased length,
cal and not the step parents are the ones to be con- whereas asymmetrical SGA indicates that the head
sidered; information about step-parenting or even circumference is spared and is normal for age; sym-
adoption may not be offered without specific ques- metrical SGA may indicate a more severe condi-
tioning. Further, it is useful to determine the grow- tion, whereas asymmetrical SGA is less severe.
ing environment of the parents to determine if their Approximately 80 % of affected SGA children will
present adult stature truly reflects their genetic catch up to the normal growth percentiles by age 2
potential: Did they grow in a war zone or were they years, but a minority remain small for their entire
deprived of adequate nutrition? growing period and attain short adult heights; the
symmetrical SGA subjects are those most likely to
Idiopathic Short Stature continue to have retarded growth. SGA patients are
ISS denotes a child in whom no other cause of characteristically thin, have bone age appropriate
short stature can be found. The cause of ISS is for chronologic age, and usually have a rate of
unknown but may be due to large effects of a few growth within the normal range for bone age. SGA
genes or small additive effects of numerous is a risk factor for a decreased age of onset of
polymorphisms. Unfortunately, many studies on puberty although many enter at the usual age range.
growth mix genetic short stature and constitutional GH treatment is approved by the FDA for those
Abnormalities of Growth 71

with SGA who do not catch up to the normal height Turner’s patients may go into puberty spontane-
distribution by 2 years of age. Numerous syn- ously and may even be fertile; variation is promi-
dromes, some discussed in this chapter, are related nent in Turner syndrome due to mosaicism.
to an SGA birth weight. Neonatal diabetes is associ- Frequent episodes of otitis media in childhood
ated with intrauterine growth restriction and SGA. may lead to conductive hearing loss, although
sensorineural hearing loss is possible, as well.
Turner Syndrome and SHOX Mutation Abnormalities of the shape of the kidneys (e.g.,
Turner syndrome is caused by a loss or abnor- horseshoe kidneys or duplications) may lead to
mality of one X chromosome or a portion of it. frequent urinary tract infections in those with
Classic cases of Turner syndrome have a 45X abnormal kidney function, so ultrasound evalua-
karyotype although variants are common. Turner tion of kidneys is indicated. Left-sided heart
syndrome has a prevalence between 1:2,000 and anomalies (such as coarctation of the aorta often
1:5,000 live births, making it a common cause of first noted due to hypertension soon after birth)
short stature (also see chapters on sexual differ- are common. Affected girls require cardiac evalu-
entiation and puberty, Chaps. 8 and 9). Physical ation for heart defects and appropriate follow-up
features of Turner syndrome include short stat- care. Even normotensive patients require regular
ure (average adult height is 143 cm), streak screening for aortic dilation every 5–10 years as
gonads (a normal complement of ova is present adults. Increased incidence of Hashimoto thyroid-
at birth, but accelerated oocyte death occurs in itis and insulin insensitivity are other features.
the postnatal period), sexual infantilism, and a Variants of Turner syndrome occur with
female phenotype. Other findings include down- abnormal X chromosome or mosaicism (e.g.,
turned edges of the mouth (leading to a “fish- XO/XX). Other variants of Turner syndrome
mouth appearance”) with retrognathia and include patients with mosaicism involving the Y
high-arched palate, ptosis and epicanthal folds, chromosome, such as XO/XY. A fluorescent in
broad shield-like chest, the appearance of wide- situ hybridization (FISH) probe for Y chromo-
spaced and hypoplastic poorly pigmented nip- some material should be performed if this situa-
ples, short webbed neck with low hairline, short tion is suspected. These patients with Y
fourth metacarpals, Madelung abnormality of the chromosome material may have dysgenetic testes
wrist (radial shortening and bowing with dorsal rather than streak gonads and are at risk for
subluxation of the distal ulna), wide carrying gonadoblastoma or malignant degeneration of
angle of the arms (cubitus valgus), knock-knee the gonad, so gonadectomy is indicated. A new
appearance (genu valgum), multiple nevi, spoon- molecular probe for Turner syndrome has been
shaped (hyperconvex) hypoplastic nails, and described for an alternative method of diagnosis.
lymphedema of the extremities, particularly in The SHOX gene (*312865 SHORT STATURE
infancy. At birth, many patients have lymph- HOMEOBOX; SHOX at Xpter-p22.32) on the
edema of the extremities and loose skin folds short arm of the X chromosome near the pseudo-
around the neck, which later transforms into the autosomal region is absent on the aberrant X
classic webbed neck and also accounts for the chromosome or if there is no second X chromo-
low-set ears; this newborn appearance of Turner some. This gene is related to the short stature of
syndrome is termed the Bonnevie–Ullrich syn- Turner syndrome and gives rise to the Madelung
drome. A tendency exists toward keloid forma- deformity of the wrist. Growth hormone (GH)
tion, which can complicate attempts to correct treatment is approved for Turner syndrome; treat-
the webbed neck surgically. A girl with Turner ment increases stature in Turner syndrome above
syndrome may not manifest all the physical find- the average adult height of 143 to 152 cm or
ings and may appear as an average short girl, so more. The age to initiate estrogen replacement is
a high index of suspicion in any short girl with- controversial; if it is offered too early and in high
out a diagnosis is indicated; rarely, girls with dosage, it will decrease the adult height, while
taller familial heights will not even be short and late start may intensify psychological problems.
hypogonadism will be the indicator. In fact, rare Recent evidence shows that low doses of estrogen
72 5 Disorders of Growth

offered at the average age of puberty will not vermilion border; high-arched palate; microgna-
affect height when used in conjunction with GH. thia; dental malocclusion; low posterior hairline;
The short stature of Turner syndrome is due to webbed neck; cystic hygroma; congenital heart
the absence of the short stature homeobox (SHOX) defect (usually of the right side rather than left-
gene. However SHOX deficiency may occur with sided heart disease as in Turner syndrome) including
dysmorphic characteristics or in otherwise normal pulmonic stenosis, septal defects, and patent ductus
children who demonstrate decreased rate of growth arteriosus; shield-like chest; pectus carinatum supe-
early in life. Leri–Weill dyschondrosteosis riorly and pectus excavatum inferiorly;
(#127300 ICD+LERI-WEILL DYSCHONDROS- hypogonadism and cryptorchidism; vertebral
TEOSIS; LWD) is an autosomal dominant condi- abnormalities; cubitus valgus; clinodactyly; brachy-
tion more often found in females with multiple dactyly; blunt fingertips; lymphedema; wooly-like
skeletal anomalies; short stature is common, but consistency of hair; articulation difficulties; and
normal stature is possible. Patients have scoliosis, 25 % have developmental delay. Fifty percent of
mesomelia, increased carrying angle, limited elbow Noonan patients have gain-of-function mutations in
mobility, bowing of the radius, dorsal subluxation the tyrosine phosphatase, nonreceptor type 11
of ulna, and short, mildly curved tibia. Patients fre- (PTPN11) gene. Noonan syndrome is considered a
quently have Madelung wrist deformity (74 % of Rasopathy disorder. While GH is FDA approved
LWD patients) as found in Turner syndrome with for Noonan syndrome, patients with this mutation
limited wrist mobility. demonstrate less growth in response to GH therapy
Langer mesomelic dysplasia (#249700 ICD+ as do those without the mutation in some studies.
LANGER MESOMELIC DYSPLASIA; LMD)
is an autosomal recessive condition also due to Russell–Silver Dwarfism Syndrome
mutations in the SHOX gene and associated with Russell–Silver dwarfism syndrome (180860
disproportionate, mesomelic short stature and the RUSSELL-SILVER SYNDROME; RSS at 7p12-
Madelung deformity of the wrist. Affected chil- p11.2) in addition to SGA is characterized by
dren have micrognathia; increased lumbar lordo- hemihypertrophy, lateral asymmetry, asymmetry
sis; short, broad ulna; spontaneous subluxation of of arms and/or legs, fifth finger clinodactyly, syn-
the distal ulna; bowed dorsolateral; short radii dactyly of toes, craniofacial disproportion, delayed
with triangularization; pyramidalization, lucency fontanel closure, triangular facies, turned-down
of distal radius on forearm X-ray; short femoral corners of the mouth, and sometimes cryptorchi-
neck; marked shortening of tibia; laterally angu- dism. On radiograph, vertebral abnormalities
lated tibia; and rudimentary fibula. including absent sacrum and absent coccyx may
be seen. It is usually sporadic but is rarely described
Syndromes of Short Stature with maternal uniparental disomy (UPD). The
main genetic defects detected are at the epigenetic
Noonan Syndrome level: hypomethylation of the imprinting control
Noonan syndrome (*163950 NOONAN region 1 (ICR1) on 11p15 in around 44 % of cases
SYNDROME 1; NS1 at 12q24) is an autosomal and maternal uniparental disomy of chromosome
dominant condition occurring in 1 in 1,000–1 in 7 (UPD(7)mat) in 5–10 % of cases. The ICR1
2,500 births that includes many features that resem- abnormality results in biallelic expression of H19
ble those of Turner syndrome and others that do not, and loss of IGF-2 expression in pathologic cells.
but does not include SGA (see Chap. 7). Some
characteristics are short stature (of postnatal onset); Fetal Alcohol Syndrome
failure to thrive in infancy; triangular, low-set poste- Fetal alcohol syndrome leads to IUGR and is
riorly rotated ears; nerve deafness; ptosis; hyper- associated with microcephaly, short palpebral
telorism; down-slanting palpebral fissures; fissure, epicanthal folds, small jaws and short
epicanthal folds; myopia; blue-green irides; philtrum of the lip, cardiac defects, and delay in
deeply grooved philtrum; high peaks of upper lip mental development.
Abnormalities of Growth 73

Syndromes of Obesity and Short hypogonadism. Spastic paraplegia, polydactyly,


Stature and other features noted in Chap. 9 are found.
Other syndromes combine short stature and obe-
sity, a combination that should awaken concern Miscellaneous Syndromes
over an organic etiology for short stature, as Numerous syndromes, some including abnormal
patients with otherwise uncomplicated obesity karyotypes, will have short stature and often SGA
are usually tall for age unless they also have con- as one of their characteristics. A complete list of
stitutional delay. such conditions is beyond the compass of this vol-
ume, so the reader is referred to a more complete
Prader–Willi Syndrome source such as Smith’s Recognizable Patterns of
The Prader–Willi syndrome (#176270 PRADER- Human Malformation in Suggested Reading.
WILLI SYNDROME; PWS—due to deletion or
maternal uniparental disomy of 15q11.2-q12) is
manifested by poor weight gain in infancy and Growth and Attention-Deficit
early childhood because of poor neonatal feeding Disorder
caused by diminished swallowing and sucking
reflexes. This condition is followed by insatiable An increasing number of children are treated with
polyphagia and rapid weight gain occurring after medications for ADHD and its variants. Data from
1 year, leading to obesity. Other features include over 40 years ago demonstrated a decreased growth
narrow bitemporal head dimension, almond- rate which may normalize after few years of treat-
shaped eyes, strabismus, myopia, thin upper lip, ment. Untreated children with ADHD appeared to
down turned corners of mouth, viscous saliva, grow at a normal rate, so the medication appears to
hypoventilation and hypoxia, hypogonadotropic be the etiology for growth diminished related to
hypogonadism (HH), cryptorchidism, small appetite suppression and reduced nutrition.
penis and scrotum, hypoplastic labia, amenor- Remarkable growth retardation associated with sig-
rhea, and limb and skeletal abnormalities includ- nificant weight loss or failure to gain weight may
ing scoliosis, kyphosis, osteopenia, small hands occur with the use of this class of medications.
and feet (acromicria), and narrow hands with a
straight ulnar border. CNS abnormalities include Chronic Disease and Malnutrition
fetal and neonatal hypotonia (such poor fetal Chronic conditions frequently affect growth,
motion leads to the assumption of fetal death by and any pediatrics textbook will have numerous
pregnant mothers in some cases), developmental etiologies on virtually every page. Chronic
delay, behavioral problems, sleep disturbances, infectious diseases causing diminished growth
hyporeflexia, articulation difficulty, skin and scab include diarrhea, respiratory tract infections,
picking, and high pain threshold. intestinal parasitic infections, malaria, HIV
GH therapy is approved for PWS, and caution infection, schistosomiasis, Cryptosporidium
in its use is important as fatal respiratory failure parvum infection, and Helicobacter pylori
has developed in some treated patients (see Chap. infection. The effects of infectious disease on
9 and below). growth may be mediated by diminished nutrition
or the effect of inflammatory cytokines.
Bardet–Biedl Syndrome A chronic disease of any organ system (cardiac,
Laurence–Moon syndrome is now combined with pulmonary, gastrointestinal, hematologic, renal,
the Bardet–Biedl (#209900 Bardet-Biedl syn- immunologic) can cause growth retardation. Indeed,
drome) syndrome with an autosomal recessive the onset of a chronic disease may be indicated by
pattern of inheritance. Obesity, short stature, an inflection point demonstrating a decrease in
developmental delay, and retinitis pigmentosum growth rate noted on the child’s growth chart; this is
can be associated with hypogonadotropic why height must be measured accurately at each
hypogonadism or alternatively hypergonadotropic visit. A few examples are highlighted below.
74 5 Disorders of Growth

Celiac disease occurs in close to 1 % of the “Failure to thrive” includes a wide variety of
general population, is more frequent in conditions in which an infant is not growing well.
Caucasians than in other groups, and 7–10 % of Usually, the weight is affected before and to a
those with type 1 diabetes mellitus have celiac greater degree than the length or height; this is in
disease with a rising incidence in the years after contrast to the effects of hypothyroidism and GH
diagnosis. Celiac disease affects growth, pubertal deficiency where weight for height is maintained
development, bone growth, and fertility and must or increased, but growth slows, leading to a chubby
always be considered in undiagnosed short stat- appearance. These infants may have as an etiology
ure. Crohn’s disease is associated with short stat- parents inexperienced in feeding infants, aberrant
ure. Juvenile arthritis also manifests with short intrafamilial dynamics, gastric reflux, and many
stature as do other autoimmune disorders with other chronic conditions found in general pediat-
growth failure. rics texts. Substantial decrease in BMI is usually
Many environmental influences (nutrition, an indication of a nutritional disease rather than an
infection, psychosocial stress, food contami- endocrine problem, although some endocrine con-
nants, pollution, and hypoxia) are considered ditions (e.g., long-standing hyperthyroidism or the
as proximate factors influencing growth which diencephalic syndrome) can indeed have decreased
interact with culture, behavior, socioeconomic weight for height. There is no reason to search for
status, social status, poverty, and political an endocrine cause of poor growth without first
economy which represent distant effects on determining if nutritional status is adequate.
growth. These proximal and distal factors are Chronic kidney disease impairs growth through
superimposed on a genetic endowment for stat- numerous factors including acidosis, decreased
ure in a manner described as genetics × envi- nutrition, and renal osteodystrophy. Growth post-
ronment. Pollutants reported to affect growth transplant improves especially if transplant occurs
are low levels of lead (below 25 μg/dL), PCBs, before 6 years of age and is increased with
polychlorinated dibenzofurans, and polychlori- decreased use of growth-suppressing immunosup-
nated and the effects of aflatoxin contamination pressive agents such as glucocorticoid therapy.
of foods. Growth rate increases with GH treatment as chronic
IUGR results from prenatal exposure to kidney disease is an FDA-approved use of GH.
cocaine, other drugs of abuse, and cigarettes as Cystinosis (#219800 ICD+ CYSTINOSIS,
well as alcohol. Environmental influences can NEPHROPATHIC; CTNS) is an autosomal
affect pubertal growth as described in Chap. 9. recessive disorder due to a mutation in the cysti-
Malnutrition, a frequent cause of short stature nosin gene (CTNS) in which there is defective
in the world, may be due to maternal neglect, pov- cystine transport from lysosomes to the cyto-
erty and lack of available food, neurologic impair- plasm. This leads to membrane-bound cystine
ment that decreases interest in food or causes crystal accumulation leading to impairment of
inability to swallow it, malabsorption, or volun- various organs, particularly kidneys where renal
tary or involuntary decrease in food intake. Serum tubular Fanconi syndrome develops causing
IGF-1 may be decreased with any of these condi- polyuria, polydipsia, dehydration, urate and cal-
tions, and the unwary diagnostician may incor- cium oxalate calculi, acidosis, and hypophos-
rectly consider that GH deficiency is at fault; in phatemic rickets. Besides impacting postnatal
states of malnutrition or starvation, GH is often growth, cystinosis is associated with primary
elevated, whereas IGF-1 is decreased, and GH hypothyroidism, insulin-dependent diabetes
administration will do no good. On the other hand, mellitus due to pancreatic insufficiency, and
obesity suppresses growth hormone secretion delayed puberty due to primary gonadal failure.
while IGF-1 levels remain normal. Malnutrition is Other features of cystinosis include impairment
the major cause of short stature in the developing of the eyes (photophobia, peripheral retinopa-
world. Parasite infestation can stunt growth even if thy, decreased visual acuity, corneal crystals,
adequate nutrition is available. recurrent corneal erosions), hepatomegaly, and
Abnormalities of Growth 75

splenomegaly. Laboratory features include pro- by GH. If adrenocorticotropic hormone (ACTH)


teinuria, glucosuria with normal blood glucose, also is absent, more profound hypoglycemia is
hyponatremia, hypokalemia, carnitine defi- possible (cortisol also stimulates gluconeogene-
ciency, hypophosphatemia, generalized amino- sis and opposes insulin action), and in either situ-
aciduria, microscopic hematuria, and elevated ation, hypoglycemic seizures may occur. The
white blood cell cystine. Various neurological combination of hypoglycemic seizures and nor-
defects develop. mal birth length without a history of complica-
tions of the birth process should alert the observer
Endocrine Causes of Short Stature of the possibility of neonatal hypopituitarism; if
the patient is a boy with microphallus, the diag-
Congenital Growth Hormone Deficiency nosis is even more likely. Midline defects ranging
Congenital GH deficiency occurs more fre- from cleft palate to encephalocele are associated
quently than one might expect as estimates range with congenital hypopituitarism. Optic hypopla-
to 1 in 3,500 children. When acquired GH defi- sia (not atrophy which occurs at an older age)
ciency (such as that caused by head and neck irra- with any degree of visual impairment, usually
diation used to treat children with cancer) is associated with pendular or horizontal nystagmus
added to the equation, the condition is even more (to and fro without any predominant direction), is
common. GH deficiency must always be consid- a classic indication of associated congenital
ered as a possible etiology in a patient with hypopituitarism. Absence of the septum pellu-
growth failure and no other identified cause. cidum is found in about 50 % of patients with
Congenital GH deficiency does not strongly optic hypoplasia and pituitary impairment, lead-
affect fetal growth, as the child has a near-normal ing to the diagnosis of septo-optic dysplasia
birth length (GH receptors are normally decreased described below. Thus, if there is diminished
in fetal life compared to postnatal life limiting the vision in association with hypoglycemia and, in a
effect of GH in fetal life), although a minor boy, microphallus, the diagnosis of hypopituita-
decrease in birth size may occur. Soon after birth, rism is more likely. If hypoglycemia is apparent
however, careful observation will indicate an and not adequately treated, developmental delay
abnormally low growth rate on careful measure- may be the outcome instead of the usually normal
ments, and plotting an accurate growth chart will intellect found in children with GH deficiency.
demonstrate this finding. The patient will be Hypoglycemia in the newborn period due to GH
obviously short for age by 2–4 years, and the or cortisol deficiency requires a lower glucose
diagnosis often will be made at this point but can infusion rate to maintain (less than 10 mg/kg/
be made much earlier with careful measurements. min) than states of insulin excess (often more
Classic GH-deficient patients will have a cheru- than 12–15 mg/kg/min) to maintain normal blood
bic appearance because of their chubbiness. They sugar (see Chap. 12).
will speak with a high-pitched voice caused by Breech delivery occurs in greater prevalence
their small larynx but have adequate intellectual in patients with congenital hypopituitarism, espe-
and vocal ability to speak appropriately for cially in boys. Any type of birth trauma may
chronologic age. They appear precocious because increase the risk of congenital hypopituitarism.
their appearance suggests a younger age, but
their speech and abilities suggest an older one. Isolated GH Deficiency
Male children may have microphallus (stretched Idiopathic, isolated GH deficiency is the most
penis length less than 2 cm, which is 2.5 standard common diagnosis. However, familial GH defi-
deviations below the mean), especially if there is ciency may occur in autosomal recessive or dom-
associated gonadotropin deficiency (see Chap. inant patterns or in an X-linked pattern due to
9). Hypoglycemia may occur because of the mutations in the gene for GH or for pituitary
absence of the gluconeogenic effects of GH and development. Decreased hypothalamic GHRH
the lack of insulin antagonism normally caused action due to a mutation of the GHRH receptor
76 5 Disorders of Growth

gene (*139191 GROWTH HORMONE- Other midline defects of the central nervous
RELEASING HORMONE RECEPTOR; system associated with pituitary deficiencies
GHRHR) is autosomal recessive (#262400 range from a simple cleft palate (6 % have growth
ICD+ISOLATED GROWTH HORMONE hormone deficiency) up to holoprosencephaly or
DEFICIENCY, TYPE IA; IGHD1A), while a anencephaly. A single maxillary incisor is associ-
loss of function mutation in the GH gene ated with pituitary dysfunction (see below).
(#612781 ISOLATED GROWTH HORMONE Rarely patients have absent anterior pituitary
DEFICIENCY, TYPE IB; IGHD1B) is autoso- glands and therefore no somatotrophs. Loss of
mal dominant. GH deficiency with hypogamma- ACTH function leads to a potentially fatal adrenal
globulinemia is inherited in an X-linked pattern failure when untreated. However, most patients
(#307200 ICD+ISOLATED GROWTH with congenital hypopituitarism are less strik-
HORMONE DEFICIENCY, TYPE III; IGHD3). ingly affected. Hypopituitarism due to a defect in
With GH treatment and therefore exposure to GH the LHX1 gene (#262600 PITUITARY
for the first time in a patient lacking the growth DWARFISM III at 9q34.3) leads to loss of GH,
hormone gene GH antibodies may develop and prolactin (Prl), luteinizing hormone (LH), and
eliminate the effects of GH treatment on growth. follicle-stimulating hormone (FSH) and thyroid-
The Growth Hormone Research Society uses stimulating hormone (TSH) in an autosomal
hypoglycemia, prolonged jaundice, microphallus, recessive pattern associated with severe restric-
or traumatic delivery and craniofacial midline tion of head rotation. Mutation of PROP-1
abnormalities as findings that should awaken con- (*601538 PROPHET OF PIT1, PAIRED-LIKE
cern about neonatal growth hormone deficiency. HOMEODOMAIN TRANSCRIPTION
FACTOR; PROP1 at 5q) causes GH, gonadotro-
Combined Pituitary Defects pin, and TSH deficiency and more rarely, ACTH
Congenital defects of the CNS may be associated deficiency, which may develop later; affected
with single hypothalamic–pituitary defects or patients must be monitored for the late onset of
more commonly multiple deficiencies. the serious additional complication of ACTH
Septo-optic dysplasia (#182230 SEPTOOPTIC deficiency. Mutations of POU1F1 (PIT *173110
DYSPLASIA at 3p21.2-p21.1, which may be due POU DOMAIN, CLASS 1, TRANSCRIPTION
to a mutation of the homeobox gene HESX1) or FACTOR 1; POU1F1 at 3p11) cause deficiency in
the appearance of small, pale optic disks is asso- GH, TSH, and Prl.
ciated with impaired vision ranging from mild
defects to blindness and is often found with hori- GH Resistance
zontal or pendular (to and fro) nystagmus. Optic GH resistance is indicated by elevated GH with
hypoplasia is to be differentiated from optic atro- decreased serum IGF-1. The first described form
phy which may be an ominous signs of a tumor of hereditary GH insensitivity, Laron dwarfism
causing the degeneration of a previously nor- (#262500 pituitary dwarfism II at 5p13-p12), is
mally formed optic disk. About 50 % of patients due to an absence of receptors for GH, and serum
with optic dysplasia have absence of the septum GHBP is low as a reflection of absent GH recep-
pellucidum. A prospective study of 47 children tors. The pattern is autosomal recessive. Other
with septo-optic dysplasia found hormone defi- forms of GH resistance have postreceptor defects,
ciencies in 71.7 % with 64.1 % of subjects having so that serum GHBP is normal. Affected patients
GHD, 48.5 % with hyperprolactinemia, 34.9 % cannot respond to GH administration with
with central hypothyroidism, 17.1 % with central increased growth or increased IGF-1 concentra-
adrenal insufficiency, and 4.3 % with central tions; patients do respond to IGF-1 administra-
diabetes insipidus (DI). There was a significant tion with increased growth, and this is the only
risk of obesity. Endocrine defects are not univer- modality that will be effective. In this syndrome,
sal but should be considered in all patients with puberty is delayed, the forehead is prominent,
septo-optic dysplasia (see Chap. 9). sclerae are blue, and dentition is delayed. This is
Abnormalities of Growth 77

an autosomal recessive condition; the GHR gene GH secretion on testing but respond to GH
is mapped to 5p13-p12 (*600946 GROWTH administration with increased growth.
HORMONE RECEPTOR; GHR). Less severe Mutations of the ALS gene (+601489
mutations of the GHR appear to lead to short stat- INSULIN-LIKE GROWTH FACTOR-BINDING
ure of a moderate nature. PROTEIN, ACID-LABILE SUBUNIT; IGFALS)
Children resistant to GH are now considered will hamper growth and delay puberty. The main
to have severe primary IGF-1 deficiency, char- genetic defects detected are at the epigenetic
acterized by unresponsiveness to GH in IGF-1- level: hypomethylation of the imprinting control
producing tissues. This is the indication for the region 1 (ICR1) on 11p15 in around 44 % of
use of IGF-1 (rhIGF-1 or mecasermin) therapy cases and maternal uniparental disomy of chro-
which is approved by the FDA for clinical use. mosome 7 (UPD(7)mat) in 5–10 % of cases.
Since some children with ISS do not have There is a poor growth response to GH treatment
excellent growth response to GH, some suggest and low levels of plasma IGF-1 and IGFBP3; the
that these may benefit from IGF-1 treatment growth failure is milder than expected with the
even without a specific FDA-approved indica- low IGF-1 and IGFBP3.
tion. Other clinical researchers suggest that the A rare cause of growth failure is a mutation in
combination of GH and IGF-1 will lead to a the insulin-like growth factor I gene (#608747
greater response than IGF-1 alone in selected INSULIN-LIKE GROWTH FACTOR I
patients. rhIGDF-1 therapy is given twice per DEFICIENCY) described in an autosomal reces-
day subcutaneously. Hypoglycemia is a risk of sive pattern. Besides growth failure, delayed bone
therapy, and a snack or meal must accompany age, and a thin appearance, other characteristics are
the dose within 20 min or the dose must be intrauterine growth restriction and SGA, micro-
omitted. If hypoglycemia persists, the dose cephaly, micrognathia, sensorineural deafness,
must be decreased. deafness, ptosis, osteopenia, clinodactyly, delayed
Pygmies (#265850 PYGMY) are individuals motor development with developmental delay in
in Africa, Philippines, and other geographic general, hyperactivity, and short attention span.
locations that have normal GH concentrations This condition should respond to IGF-1 therapy.
and normal IGF-1 but very low IGF-2 concentra- Another rare condition in children with
tions; defects in the functioning of IGF-1 recep- growth failure is a mutation of the IGF-1 receptor
tors are reported to be major influences on their (#270450 INSULIN-LIKE GROWTH FACTOR I,
poor growth in some. Because the population is RESISTANCE TO) found in an autosomal domi-
described with nutritional deficiencies as well as nant or autosomal recessive pattern. Characteristic
the genetic defect, multiple causes of short stat- features besides growth failure, delayed bone age,
ure may exist, but IGF-1 receptor defects are and a thin appearance are intrauterine growth retar-
found by some but not all investigators. dation and SGA, microcephaly, facial abnormali-
Biologically inactive GH (#262650 KOWARSKI ties including receding hairline, long smooth
SYNDROME or biologically inactive GH. philtrum, broad nasal bridge, thin upper lip, fleshy
PITUITARY DWARFISM IV) is postulated as a lower lip as well as pectus excavatum, wide-set
cause of growth failure with normal serum GH nipples, clinodactyly, delayed motor development,
concentrations as measured by radioimmunoas- speech delay, anxious affect, obsessive tendencies,
say but poor growth; growth rate increases with and agitation. Insulin-like growth factor 1 values
administration of exogenous GH, even though are elevated with growth hormone values either
the endogenous GH appears ineffective. The normal or elevated.
prevalence of affected patients is unknown; auto-
somal recessive inheritance is suggested. Partial Acquired GH Deficiency
GH deficiency appears to be a more common Abnormalities of the central nervous system.
explanation for this condition in many cases and These conditions can be devastating or fatal and
is represented by subjects who have adequate are prime examples of why the diagnosis of the
78 5 Disorders of Growth

etiology of acquired short stature must be care- Other Conditions


fully established. Abnormalities of puberty may Histiocytosis X may be associated with diabetes
be caused by many of these same conditions so insipidus, but other hypothalamic hormones may
making diagnosis equally important when puberty also be deficient and HH can occur. Granulomas
is affected (longer discussion found in Chap. 9). of tuberculosis, rare nowadays, or sarcoidosis,
Central nervous system tumors. Acquired growth postinfectious inflammation, or vascular lesions
failure is an ominous sign since it may be due to of the central nervous system can impair hypo-
a CNS tumor. If both anterior and posterior pitu- thalamic pituitary function. CNS trauma due to
itary deficiencies are noted, a central nervous accidents, child abuse, or surgery for tumors near
system tumor should be even more highly sus- the area can also affect hypothalamic pituitary
pected. To reemphasize, late onset of growth gonadal function. Hydrocephalus can cause GH
failure or a cessation of pubertal development deficiency as may any other etiology of increased
that has already started is of great concern. intracranial pressure.
Oncofertility is a new field growing out of the
Central Nervous System Tumors increasing number of cancer survivors following
Craniopharyngiomas occur with a peak incidence successful treatment who show late effects of
between 6 and 14 years of age. Patients may be treatment. Radiation treatment may cause growth
asymptomatic or symptomatic with complaints of hormone deficiency or gonadotropin deficiency
headache, visual loss, polyuria, and polydipsia. but also, when used to treat a central nervous sys-
Signs on physical exam may be either short stature tem tumor, may lead to a combination of preco-
or cessation of previously normal growth, findings cious puberty and growth hormone deficiency.
of hypothyroidism, and sexual infantilism. The growth hormone deficiency will not be dem-
Papilledema or optic atrophy (not congenital optic onstrated by extremely slow growth if the puberty
dysplasia) might be found as well. Pituitary calcifi- is progressing rapidly as the sex steroid secretion
cations may be found on a radiograph taken for stimulates growth masking the GH deficiency
other reasons such as orthodontic evaluation. CT (read below). Long-term follow-up of children
scans will show flecks of calcium within the tuber treated with CNS irradiation for leukemia dem-
cinereum in more than 80 % of cases. MRI scans onstrated atrophy of the pituitary gland or empty
before and after gadolinium will define the shape sella 10–40 years later.
and size of the tumor better although it will not
show calcification unless susceptibility weighted Psychosocial Dwarfism
imaging is used. This tumor might be cystic, and Psychosocial dwarfism (also known as psychoso-
the sella turcica may be eroded by expansion of the cial short stature and sometimes grouped with
tumor. Transsphenoidal microsurgery can be used hyperphagic short stature) may occur in one child
for extirpation if the tumor is intracellular. Larger among several other normal siblings. Abnormal
tumors that cannot be completely removed by this parent–child interaction leads to functional hypo-
method may be treated with radiation as the tumors pituitarism with GH deficiency on provocative
are radiosensitive with a combination of surgery testing; GH secretion will revert to normal within
and radiation usually recommended. Late effects days after removal from the offending situation.
of radiation are possible with CNS irradiation, Affected children have a pot-bellied, immature
however, such as secondary tumors of local tissue. appearance. They may exhibit aberrant behavior
Other tumors of the central nervous system such as drinking from toilet bowls, begging for
reside outside of the sella turcica such as germino- food from neighbors, and foraging for food in
mas of the pineal, which by their mass may affect garbage cans, in spite of apparently acceptable
growth. Astrocytomas and gliomas (which may or caloric intake at home. These patients may have
may not be associated with neurofibromatosis) are had psychological abuse but rarely have signs of
other CNS tumors that may cause growth failure. physical abuse. When removed from their homes,
Abnormalities of Growth 79

Complete history and physical with careful measurement of height and weight

height is below the 3rd percentile on the CDC growth charts or is below the 5th percentile for corrected midparental height or growth rate is below the 5th percentile
Yes No

BMI is low probably normal variation; reassurance is best approach


No Yes

historical feautures of chronic disease or malnutrition evaluate for nutritional problem before embarking upon endocrine evaluation
No Yes (although some endocrine conditions such as diencephalic syndrom may still be possible)

physical features compatible with syndrome of chondrodystrophy


No Yes
chemistry panel, cbc, sed rate normal
Yes No

free T4 and TSH normal calcium low and phosphorous high


Yes No Yes No

history or celiac panel compatible with celiac disease evaluate for hypothyroidism evaluate appropriately
No Yes
evaluate for pseudohypoparathyroidism by measuring serum PTH
IGF-1 and IGFBP3 normal
GI consult with possible small bowel biopsy
No Yes

re-evaluate nutritional status with dietary evaluation


adequate nutrition abnormal

perform GH stimulation test rectify


peak is low normal

GH deficiency; still possibly responsive to GH; individualize approach


likely response
to GH treatments MRI of Hypothalamus ⁄ pituitary to search for etiology

Fig. 5.12 The basic evaluation of short stature. Referred to the text for suggestions of detailed diagnostic procedures
and gene defects

such children may exhibit catch-up growth in effective than wished. Some patients respond to
foster homes or in the hospital, environments GH treatment without demonstrating classic GH
where one would not expect a child to thrive. deficiency; most of these more subtle indications
Even if psychotherapy is administered for pro- are beyond the range of this text, as all short stat-
longed periods, the outcome is uncertain. Growth ure conditions requiring GH treatment are best
hormone is not an appropriate treatment for psy- diagnosed and certainly treated by a pediatric
chosocial dwarfism however. endocrinologist.
Maternal deprivation (perhaps a sexist term, Classic GH deficiency is defined by the inabil-
but as the mother usually is responsible for feed- ity to increase GH concentration above an
ing the child, an often-used term) is the diagnosis accepted limit, generally 10 ng/mL in most labo-
applied to infants with poor growth rates due to ratories (although certain assays are more sensi-
parental neglect. These children may have actual tive, and lower cutoff of normal levels is used)
caloric deprivation in addition to psychological after two stimulatory tests. This limit is arbitrary
neglect. The term also is applied to situations in and is higher than values used in previous
which the mother is inexperienced in child- decades. Further, prepubertal children may not
feeding practices in which education can provide respond as well as pubertal children to GH stimu-
marked improvement. latory tests, so that different standards may be
These conditions are often grouped into the useful at different ages (according to published
term “nonorganic failure to thrive.” studies), but such standards for stimulated tests
are not available from commercial laboratories.
Diagnosis of Growth Hormone Deficiency To standardize the responses before and after
(Fig. 5.12) puberty, some investigators suggest priming the
The diagnosis of GH deficiency has become prepubertal child with a dose of estrogen or pro-
more complex with increased knowledge of GH pranolol (propranolol can precipitate hypoglyce-
physiology. The dividing line between classic, mia and should not be used if hypoglycemia
severe GH deficiency and normal GH secretion is (or asthma) is already present). As mentioned,
blurry as our classic tests have proven less the measurement of a single sample for basal GH
80 5 Disorders of Growth

concentrations is useless in the diagnosis of GH cian during the test. The patient will be extremely
deficiency but may be helpful in GH resistance. uncomfortable during the hypoglycemia that
Secretagogues invoked in the testing for GH results. Glucose and GH should be measured at 0,
deficiency are given early in the morning after an 15, 20, 30, 60, and 90 min. Cortisol may be mea-
overnight fast (hypoglycemic patients should not sured at 60 and 90 min if this test is performed to
be forced to fast unobserved, so the test must be test ACTH reserve as well as GH secretion. If
modified!). Two tests are usually performed severe hypoglycemia occurs, dextrose must be
because 30–50 % of normal children can fail to administered immediately; no greater concentra-
increase GH concentration after one test; how- tion than 25 % dextrose at 1 mL/kg should be
ever, two tests yield a false positive (failure of a infused so that blood sugar will increase but no
normal child to reach the normal limit of serum severe change in osmolality will occur. Growth
GH) more than 10 % of the time. Hypothyroid hormone-releasing factor (GHRH) is not com-
individuals will not respond normally to GH mercially available for testing and offers little
secretagogues, and hypothyroidism is much more improvement in testing over the other regimens.
common than GH deficiency; thyroid status must (When it was administered in a dose of 5–10 μg/
be tested before GH testing. kg, serum GH concentrations increased in normal
Several pharmacologic tests are commonly individuals.) GH-deficient patients demonstrate
invoked as secretagogues. L-Dopa is used in doses either no increase in GH or a blunted increase to
of 125 mg for body weight up to 15 kg, 250 mg secretagogues. Those with a pituitary defect have
for weight up to 35 kg, and 500 mg for body no response, whereas those with a hypothalamic
weight over 35 kg; samples are taken at 0, 30, 60, defect may respond partially.
and 90 min. Nausea and vomiting are possible Nonpharmacologic tests are available in addi-
side effects for a few hours after administration. tion to the stimulatory tests. After 10 min of vig-
Clonidine in a dose of 0.1–0.15 mg/m2 can be orous exercise, normal children will increase
given and samples obtained at the same time their GH in 80 % of trials. Sequential serum
sequenced; side effects are possible hypotension samples may be taken at night in the hope that
and lethargy for several hours, so we prefer the the peak that customarily occurs 90 min after the
lower range of dosage. Since it may be dangerous onset of sleep may be captured, but this method is
to have emesis while sleeping, usually the L-dopa expensive and inconvenient. Sampling for GH
test is performed before the clonidine test. levels every 10–15 min for 24 or 12 h has been
Intravenous arginine infusion of 0.5 g/kg body suggested as a way to determine whether a nor-
weight up to 20 g over a 20-min period can be mal circadian rhythm of GH secretion is present;
given with samples taken at the same time peri- the facilities for such a study are generally
ods as mentioned earlier; no side effects are unavailable.
likely. A glucagon stimulation test may be accom- Due to remarkable lack of reliability in labora-
plished with the IM administration of a dose of 30 tory testing of GH secretion, results must be
μg/kg glucagon (maximum 1 mg) and collecting interpreted in view of anthropomorphic growth
GH samples at 0, 30, 60, 90, 120, 150, and 180 data to make the diagnosis of GH deficiency. The
min afterwards; nausea and hyperglycemia are addition of measurement of IGF-1 improves
the side effects. Insulin-induced hypoglycemia accuracy however.
(0.075–0.1 U/kg body weight of insulin given Plasma IGF-1 concentrations must be inter-
i.v.) is an effective but extremely dangerous test; preted with caution. Low values may mean mal-
the patient must be shown to have a normal fast- nutrition rather than GH deficiency; in
ing blood sugar just before the test and must not malnutrition, GH values usually increase, and
have a known tendency toward hypoglycemia, a IGF-1 values decrease because GH receptors
working i.v. line must be available to infuse dex- decrease. In delayed puberty, values are more
trose should a hypoglycemic seizure occur, and appropriate for bone age than for chronologic age
the patient must be watched carefully by a physi- and may be incorrectly interpreted as abnormally
Abnormalities of Growth 81

low in such a situation; thus a 14-year-old with an available. A new long-acting preparation is said to
11-year-old bone age who has an IGF-1 value be available soon.
inappropriate for a 14-year-old but appropriate GH is presently indicated for GH-deficient
for an 11-year-old is likely normal. While IGF-1 patients, Turner syndrome (see Chaps. 8 and 9),
assays can be helpful, their accuracy is called into growth failure of renal insufficiency, Noonan syn-
question. The measurement of IGF-BP3 in addi- drome, SHOX gene deletion either in Leri–Weill
tion to IGF-1 may add accuracy to the diagnosis syndrome with one missing SHOX allele or
of GH deficiency especially in those under 3 Langer syndrome, SGA, and uncomplicated ISS.
years or in malnutrition. GH is approved for use in children with Prader–
Willi syndrome for short stature, but it is the meta-
Growth Hormone Treatment of GH Deficiency bolic effects and improvement of muscle tone in
and Other Disorders Approved for GH which it exerts its most important effects; respira-
Treatment tory compromise however is a contraindication to
The treatment of GH deficiency is accomplished GH therapy as GH can worsen respiratory obstruc-
by the use of biosynthetic recombinant DNA- tion through soft tissue enlargement in these chil-
derived GH. Previously, cadaver-donated dren, and only after establishing adequate
pituitary-derived GH was administered, but the respiratory function should GH be initiated in
recognition that some batches were contaminated Prader Wili. Since respiratory fatalities are more
with the prions of Creutzfeldt–Jakob disease likely in the first 9 months of treatment, it is rec-
(CJD) led to the discontinuation of such therapy ommended that the dose of GH be started at about
in 1983–1985 and the switch to recombinant- ½ the standard dose and increased over the first
derived GH; it is possible that some children few months.
treated before that date will still develop CJD as GH is used illicitly in numerous ways. Athletes
cases were reported as late as 2014. Biosynthetic and weight lifters obtain GH by unscrupulous
GH is available with the 191-amino acid sequence providers to increase muscle strength, but no ben-
of the natural GH molecule from many compa- efit has been proven in these conditions (at least
nies. Daily subcutaneous injections are adminis- with standard doses which are likely considerably
tered in doses starting at about 0.18–0.3 mg/kg/ lower than illicit doses). GH treatment of adults
week given 6 or 7 days/week; about twice this with severe GH deficiency is indicated to main-
dose is approved during puberty as there is higher tain bone density and improve skin, muscle con-
endogenous GH release during puberty in normal dition mood, and memory. While GH is indicated
individuals. Dosage is prescribed related to in the treatment of adults with bona fide GH defi-
weight and titrated to the growth response. The ciency, the use of GH in the general treatment of
aim is also to maintain plasma IGF-1 values in aging without a diagnosis of GH deficiency is
the normal range for safer dosing and to theoreti- used in unscrupulous antiaging clinics. The
cally reduce the risk of development of neoplasm Growth Hormone Research Society recently
or other comorbidities from this mitogenic medi- reviewed all data and stated, “the clinical use of
cation. IGF-1 values are expressed in terms of Z GH or GHS (growth hormone secretagogues) in
scores which indicate the number of standard older adults, alone or in combination with testos-
deviations above or below the mean. It is sug- terone, cannot be recommended.”
gested that if IGF-1 concentrations are consis- The FDA approved GH for therapy in children
tently more than a 2.5 z score or SDS above the with ISS, and a height SDS −2.25 (<1.2 %) that is
mean, GH dose should be decreased. associated with a growth rate unlikely to permit
Syringe-and-needle administration of either attainment of adult height in the normal range, in
premixed solutions of GH or lyophilized prepara- pediatric patients whose epiphyses are not closed
tions that must be reconstituted has been replaced and for whom diagnostic evaluation excludes
by the use of mechanical pens that use cartridges other causes associated with short stature that
of premixed GH, and even needleless devices are could be observed or treated by other means.
82 5 Disorders of Growth

Consensus guidelines state that this can be decreases GH levels in basal and stimulated states,
considered if the height ranges from −2 SDS to leading to a suggestion of true GH deficiency.
−3 SDS with best effects if GH is started between Obesity is related to type 2 diabetes in genetically
5 years and early puberty. Randomized control susceptible individuals; the evaluation of an obese
studies are scarce, but a recent review summarized patient appearing to have coexisting GH deficiency
that with a mean age at start of treatment of 10 and type 2 diabetes may be difficult although most
years, mean duration of GH therapy was 5.4 years children with obesity will tend to have taller-than-
(average duration of treatment 4–7 years); the average stature so the question may not arise.
mean difference between treated and untreated GH therapy in Turner syndrome utilizes a
children was approximately 5.5 cm (3.5–7.5 cm). higher dose (0.375 mg/kg/week) than in GH defi-
The mean adult height achieved after GH therapy ciency (up to 0.3 mg/kg/week in prepuberty).
at conventional dosages was −1.5 SDS. Better Treatment may be more beneficial if started before
results occurred with early age at onset of ther- 2 years of age. Oxandrolone, a non-aromatizable
apy, higher GH dose, greater birth length, and androgen (0.05 mg/kg/day), is reported to increase
more delay in bone age. Also, children whose adult height, but liver function must be monitored
parents were of normal height gained more than and observation for virilization is necessary
those with familial short stature. The mean adult according to a study in toddlers. Concerns arose
height was increased 0.7 SD in treated children. that while estradiol administration may improve
Often the referral for short stature occurs too late, age-appropriate appearance, it may decrease adult
when the effect of GH is far less since the bone height. Data from a multidecade study of low-
age is more advanced. It is important to realize dose estrogen started at age 12 combined with GH
that with annual costs of $20,000–40,000 per started as young as 5 years suggests that this com-
year for growth hormone, a mean gain of 1 in. bination may increase growth rate safely. Side
means an expenditure of $52634. Some may effects of GH therapy are reported at a higher
wonder if the child might be happier with the prevalence in Turner syndrome including type 2
money rather than the treatment. Indeed there is diabetes mellitus and scoliosis, so close observa-
preliminary data suggesting that children with tion is necessary.
ISS and GHD become progressively less happy There are studies of the use of GnRH agonists
with treatment and clinic visits compared to typi- to delay pubertal development and thereby allow
cal untreated children. the period of growth to continue; some studies use
Complications of GH treatment are rare but combined GH and GnRH. An evidence-based
include a tendency toward slipped capital femoral review found no convincing evidence that this
epiphyses, and patients must be asked about hip treatment is effective and it is not recommended.
and leg pain or the development of a waddling gait; Aromatase inhibitors stop the conversion of
the treatment is surgical. Pseudotumor cerebri is androgens to estrogens by aromatase, and it is
rare but may cause a severe headache along with estrogen that advances bone age and ultimately
papilledema: although it should be reversible, if it causes cessation of growth. Letrozole and anas-
is allowed to continue, impairment or loss of vision trozole have been successfully used in boys with
may result from effects on the optic nerve. constitutional delay to allow a taller adult height
Hypothyroidism may develop during GH therapy, achievement. Testes may become much larger,
and a free T4 and TSH value should be obtained and testosterone may rise substantially during
annually or if growth rate decreases. Development therapy. Side effects are few but may include
of antibodies to GH is less a problem with the use abnormalities in the shape of vertebrae.
of human sequence GH but should be considered if Theoretically, adult bone density could be
a patient ceases to grow as expected while receiv- decreased, but there is no proof of this concern at
ing therapy with GH. Type 2 diabetes is theoreti- present. This is not an approved therapy and must
cally possible due to the insulin resistance caused be considered experimental and only to be used in
by GH therapy but rarely occurs. Indeed, obesity controlled studies at present.
Abnormalities of Growth 83

Recently, French researchers released data sports that will realistically allow success and
from a large ongoing prospective study and foster self-confidence; for example, soccer is
reported increased prevalence of bone cancer and more appropriate than basketball, and martial arts
cardiovascular and CNS vascular disorders in classes can lead to greater self-confidence in an
young adults previously treated with GH as teen- insecure shorter child. The ultimate psychologi-
agers and younger. Review of the records of other cal outlook of a short child may result, in large
European countries showed no such findings, so part, from the manner in which the child was
the explanation for this unusual set of data was approached by family, teachers, peers, and physi-
not clear, and reanalysis of the statistics indicated cians during the early years.
that these conclusions cannot be supported. At Children diagnosed with GH deficiency may
the time of this writing, a second paper from this later develop other pituitary deficiencies such as
group linking GH treatment with hemorrhagic ACTH deficiency even if it is not found at initial
stroke in early adulthood was released. While it is evaluation. Thus every child with diagnosed GH
considered a good study, methodological issues deficiency should be monitored at intervals for
are raised, but the cause and effect of GH treat- ACTH deficiency. Likewise, GH treated children
ment and stroke remain to be established. It is should be monitored for the development of
recommended that patients and families be noti- hypothyroidism annually.
fied of these possibilities as they decide upon
therapy. The FDA is presently investigating these Other Endocrine Disorders
data, but findings have not changed clinical prac- Other Endocrine or Syndromic
tice in the USA and the FDA has not suggested Conditions
GH use change from the standard manner.
Hypothyroidism
Controversy exists over the negative psycho-
logical effects of short stature. There is a great Hypothyroidism will decrease the growth rate
need for robust scientific study of these effects and retard skeletal development. Most children
rather than relying on common stereotypes that with congenital hypothyroidism will be diag-
short stature diminishes quality of life. Further, it nosed with neonatal screening procedures, but
is the patients who are brought for evaluation acquired hypothyroidism is still a frequent occur-
who are usually studied rather than the children rence in later childhood and adolescence (see
in the community with short stature who do not Chap. 6). It is quite unfortunate if a hypothyroid
come to medical attention. Nonetheless, psycho- patient who can be easily treated with inexpen-
logical support should be least considered for a sive pills is considered inappropriately for GH
short child whether or not medical therapy is therapy, but this author has seen this situation.
offered. Parents should be counseled against
thinking that the fact that their child is shorter Cushing Syndrome
than average is a tragedy and that therapy is Cushing syndrome refers to excess glucocorti-
required at all costs. In view of the constant bom- coid exposure from endogenous or exogenous
bardment by the media that leadership, friend- sources. Topical and even inhaled preparations of
ship, and indeed the company of the opposite sex glucocorticoid can retard growth and must be
require tall stature (at least in boys), some boys considered a potential cause of decreased growth
(and some girls) may become depressed; the (see Chap. 10). The decreased growth in Cushing
most severe of these tendencies are rare, but the syndrome is generally considered due to sup-
physician must be sensitive to such a possibility. pressed growth hormone secretion through
If the child’s parents are shorter than average and increased somatostatin secretion.
unhappy about it, they may tend to read into the
child’s future the unpleasant experiences they Pseudohypoparathyroidism
had, which may not, after all, directly reflect on Pseudohypoparathyroidism type 1A (#103580
their height. Children should be counseled toward ICD+PSEUDOHYPOPARATHYROIDISM,
84 5 Disorders of Growth

TYPE IA; PHP1A) with resistance to PTH is 3. Height corrected for midparental height of
accompanied by Albright’s hereditary osteodys- <−2 SD or <2.3 % (the Growth Hormone
trophy, a chubby appearance, short stature, short Research Society uses <1.5 SD)
fourth metacarpals, round face, and developmen- 4. Signs indicative of an intracranial lesion
tal delay. Hypocalcemia and hyperphosphatemia including a tumor or trauma
can be treated with 1,25 OH vitamin D therapy, 5. Signs of multiple pituitary hormone defi-
but the poor growth will remain (see Chap. 7). ciency (MPHD)
Some patients demonstrate GHRH resistance. 6. Neonatal symptoms and signs of GHD
7. A history of central nervous system radiation
Rickets
Rickets is making a comeback in the developed A combination of two or more of these charac-
world due to lack of or inconsistent vitamin D teristics warrants increased concern (Table 5.2).
supplementation of breast-fed infants (there is A full history and physical examination are
inadequate vitamin D in breast milk) who are essential for the evaluation of short stature to
shielded from sunlight (e.g., in the developing reveal genetic or nutritional influences or sys-
world due to cultural practices that cover the skin temic diseases. Interfamilial interactions can be
or the use of sunscreen to avoid skin cancer); observed during the interview process to evaluate
growth is impaired in nutritional rickets. the possibility of psychosocial dwarfism. If no
Hypophosphatemic rickets is associated with previous height measurements are available, a
short stature, and GH treatment was investigated history of changes in shoe or clothing size can be
but not shown to be effective to increase growth of value to determine whether the child is grow-
(see Chap. 7). The many genetic forms of rickets ing adequately, although careful follow-up mea-
are mostly associated with short stature. Return surements over at least 3 months (and longer if
to metabolic balance in some forms improves possible) are necessary for diagnosis.
growth rate.
History and Physical Examination
Diabetes Mellitus In many cases, the diagnosis is apparent from the
Modern treatment of diabetes mellitus offers medical history. The clinician must query for
excellent control, but not all patients adhere to abnormalities or toxic exposures of pregnancy,
recommended treatment. Growth can be affected nutritional status during pregnancy, difficulties of
by poor control. In the most severe cases, poor delivery, and degree of prematurity, if any, and
control leads to Mauriac syndrome with atten- evaluate weight for gestational age (Fig. 5.1).
dant short stature, delayed pubertal development, Determination of age of puberty of parents (men-
and hepatomegaly with abnormal liver function arche in mothers and age of physical changes of
(see Chap. 11). puberty or cessation of growth in fathers) or sib-
lings may suggest a tendency toward constitu-
Diagnosis of Short Stature tional delay in growth or adolescence. Family
As stated above, the following criteria may be history of chronic disease, including neurologic or
used as indicators of the need for an evaluation endocrine conditions, and the child’s history of
for disorders of growth: symptoms of chronic disease of almost any organ
can be of great importance, as most chronic dis-
1. Stature <2.25 SD or 1.2 % (the Pediatric eases can decrease growth. Remember that virtu-
Endocrine Society uses <2 SD below the mean ally every page of a textbook of pediatrics discusses
for age and gender or 2.3 % for ISS, while the a condition which can impair growth.
Growth Hormone Research Society uses <3 SD Physical examination must be complete to
below the mean) search for abnormalities of nutrition, chronic
2. Height velocity <2 SD below mean for bone disease, and neurologic problems, especially of
age the central nervous system (CNS). Physical
Abnormalities of Growth 85

Table 5.2 The evaluation of short stature


Test Rationale
CBC Anemia: nutritional, chronic disease, malignancy
Leukocytosis: inflammation, infection
Leukopenia: bone marrow failure syndromes
Thrombocytopenia: malignancy, infection
ESR, CRP Inflammation of infection, inflammatory diseases,
malignancy
Chemistry panel (electrolytes, liver enzymes, BUN) Signs of acute or chronic hepatic, renal, adrenal
dysfunction; hydration and acid-base status
Carotene, folate, tissue trans-glutaminase antibody, or reflexed Assess malabsorption; detect celiac disease
celiac panel
Urinalysis Signs of renal dysfunction, hydration; renal
tubular acidosis
Karyotype, candidate gene analysis, SNP analysis using Evaluate for genetic syndromes
microarrays on DNA chips, array-comparative genomic
hybridization (array-CGH), or other genomic techniques
Cranial MRI imaging Assesses hypothalamic-pituitary tumors
(craniopharyngioma, glioma, germinoma) or
congenital midline defects
Bone age Determine physiologic maturation, and evaluate
height potential
IGF-1, IGF BP3 Reflects growth hormone status or nutrition
Free thyroxine and TSH Detects hypothyroidism
Prolactin Elevated in hypothalamic dysfunction or
destruction, suppressed in pituitary disease
BUN blood urea nitrogen, CBC complete blood count, CRP C-reactive protein, ESR erythrocyte sedimentation rate, IGF
BP3 insulin-like growth factor-binding protein 3, MRI magnetic resonance imaging, TSH thyroid-stimulating hormone

examination begins with the accurate measure- nutrition appears to be the problem, an endocrine
ment of height, weight, head circumference, arm condition is less likely, and other causes must be
span, upper-to-lower segment ratio, and vital considered.
signs. Height and height velocity are plotted on If no diagnosis is obvious from history or phys-
appropriate charts (see Figs. 5.4 and 5.5). It is ical examination, laboratory evaluation is per-
essential to always calculate and plot body mass formed. A screening chemistry panel for liver and
index (BMI charts found at www.CDC.com; see kidney function, a bicarbonate and electrolyte
Chap. 12); low BMI for age might indicate mal- determination to evaluate the possibility of renal
nutrition due to chronic disease, whereas elevated tubular acidosis or a metabolic acidosis syndrome,
BMI for age might raise a concern since many a complete blood count with differential, and a uri-
children with obesity have increased stature nalysis including microscopic examination may
while endocrine conditions and various syn- indicate a problem not obvious from the physical
dromes can lead to obesity and short stature. As a examination. A free thyroxine determination and
general rule, endocrine problems slowing growth TSH also are obtained to determine whether hypo-
tend to be found in heavier (but not necessarily thyroidism is present. A celiac reflexive panel
obese) children rather than thin ones with the which ensures adequate IGA to ensure validity of
possible exception of hyperthyroidism or the test will determine if unexpected celiac disease
untreated type 1 diabetes mellitus. Signs of dis- is the etiology of the problem; evaluation of caro-
ease or syndromes are evaluated. If decreased tene or folate for determination of fat absorption
86 5 Disorders of Growth

may be considered. A sedimentation rate is used Table 5.3 Etiologies of tall stature
as a general indicator of inflammation. A bone-age Variations of normal
determination will not lead to a diagnosis (although Constitutional
it may support several potential diagnoses), but Genetic
will indicate the amount of remaining growth, in a Exogenous obesity
general manner. If obesity complicates short stat- Endocrine disorders
ure and Cushing syndrome is under consideration Pituitary gigantism
(rare), a urinary free cortisol determination is Sexual precocity
obtained or an AM plasma or salivary cortisol after Thyrotoxicosis
midnight dexamethasone suppression (see Chap. Beckwith–Wiedemann syndrome
10). An elevated serum Prl concentration will sug- Infant of a diabetic mother
gest hypothalamic deficiency and lack of dopa- Nonendocrine disorders
mine suppression; if it is low, it will suggest Marfan syndrome
pituitary disease. Klinefelter syndrome
If neurologic symptoms are noted or if a CNS XYY syndrome
tumor is in the differential diagnosis, an MRI eval- Cerebral gigantism (Sotos syndrome)
Homocystinuria
uation with gadolinium contrast of the hypotha-
Weaver-Smith syndrome
lamic–pituitary area is in order. It is essential that
thin cuts be ordered and taken through the hypo- Modified from Styne DM. Growth disorder. In Greenspan
FS, Gardner DG, eds. Basic and clinical endocrinology.
thalamic–pituitary area on the MRI so that a small New York: Lange, 2011: with permission
tumor or congenital abnormality is not missed.
Suspected congenital GH deficiency also
requires an MRI to determine possible CNS Tall Stature
developmental abnormalities. Small or absent
pituitary glands, ectopic pituitary location, and Nonendocrine Causes of Tall Stature
interruption of the pituitary stalk are found in Constitutional or genetic tall stature is the other side
some congenital abnormalities associated with of the coin from constitutional or genetic short
pituitary dysfunction. stature, although it is far more rarely evaluated
Evaluation of GH secretion is usually per- (Table 5.3). Patients are taller than average, may
formed if no other diagnosis is apparent from the have a moderately advanced bone age but a height
evaluation. As mentioned earlier, IGF-1 determi- velocity appropriate for bone age, and have no
nations are preferred by many pediatric endocri- sign of the disorders described later. Puberty will
nologists rather than GH stimulatory testing. begin early in children with constitutional tall
IGF-1 values must be interpreted by pubertal and stature, so that adult height will not likely be
nutritional status and are best expressed in z above normal range, although height during
scores based upon standard deviations from the childhood was greater than normal. Obesity will
mean; +/- 2 SD are considered within the normal advance bone age and physiologic development
range. Insurance companies may insist on GH and often leads to tall stature in childhood but does
secretory testing nonetheless. An IGF-1 concen- not translate into a taller adult height.
tration may be falsely low because of nutritional Genetic tall stature occurs in a family with one
deficiency or constitutional delay, but if the or, more often, two parents taller than the normal
IGF-1 and IGF-BP 3 are normal, classic GH defi- adult range. The child can be expected to reach
ciency is less likely. If stimulation is performed, a taller-than-average height as an adult. The
the diagnosis of classic GH deficiency is rela- adjusted midparental height will help in the eval-
tively straightforward, as GH values will be uation of genetic tall stature (Fig. 5.8).
lower than the limits stated above. The other, Girls more often will be concerned about
more subtle conditions of impaired GH secretion being taller than average than will boys; often
will be more difficult to correctly diagnose. fears are intensified by parental concerns about
Abnormalities of Growth 87

tall stature and effects on social development. as postnatal increased stature and advanced bone
However, such fears may have decreased in view age but no acceleration of growth velocity, as seen
of the modern success of tall fashion models, in Soto syndrome. It is usually sporadic. Facial fea-
actresses, and athletes. Thus treatment is rarely tures include macrocephaly, large bifrontal diame-
invoked today because of ethical issues, but high- ter, flattened occiput, long philtrum, round face in
dose estrogen therapy has been used in the past infancy, retrognathia, prominent chin with central
and has been effective in slowing longitudinal dimple, large ears, strabismus, hypertelorism, epi-
growth while advancing puberty, causing a rapid canthal folds, down-slanting palpebral fissures, and
increase in bone age and thereby limiting adult depressed nasal bridge. Numerous skeletal abnor-
height in girls. Because of theoretic risks of high- malities include scoliosis, kyphosis, coxa valga,
dose estrogen therapy—such as thrombotic limited elbow extension, limited knee extension,
effects, the development of ovarian cysts, and flared metaphyses (especially distal femora and
future menstrual disorders—therapy, if ever con- humeri), camptodactyly, broad thumbs, prominent
sidered, is not to be undertaken without a careful fingertip pads, large hands, clinodactyly, talipes
description of the possible side effects. Further, equinovarus, calcaneovalgus, metatarsus adductus,
the child must be included in discussions con- short fourth metatarsals, prominent toe pads, pes
cerning her adult height. In rare circumstances, a cavus, and overriding toes. Behavior and learning
girl with a predicted height of more than 6 ft, disorders, seizures and hypotonia, and abnormali-
who is age 10–12 years with a bone age no older ties of CNS anatomy occur in some.
than 10–11 years, may hope to realize a decrease Marfan syndrome (#154700 MARFAN
in her final height of up to 2 in. with the use of SYNDROME; MFS at 15q21.1) includes dispro-
estrogen therapy. As always, a concern for hema- portionate tall stature with long-bone overgrowth
tologic disorder arises when estrogen is used, (dolichostenomelia), arm span exceeding height,
and a search for factor V Leiden deficiency is a very low upper-to-lower segment ratio, arach-
appropriate in such therapy. This therapy is not nodactyly (or long, thin fingers and toes), hyper-
recommended by the author but is presented for extensibility of the joints, as well as joint
completeness and for historical interest. contractures. Other skeletal anomalies include
Boys rarely are concerned about excessive scoliosis, kyphoscoliosis, thoracic lordosis, spon-
adult height, but testosterone therapy has been dylolisthesis, lumbosacral dural ectasia, pectus
used to precipitate pubertal development and excavatum, pectus carinatum, and pectus asym-
limit adult height. Such therapy is not recom- metric deformity. Optic anomalies include sub-
mended either. luxation of the lens (ectopia lentis) with myopia,
Cerebral gigantism (*117550 CEREBRAL corneal flatness, retinal detachment, iris hypopla-
GIGANTISM at 5q35) or Soto syndrome is rec- sia, early glaucoma, early cataracts, and down-
ognized by the prominent forehead and sharp slanting palpebral fissures. In addition, the face is
chin, with prognathism, high-arched palate, long and narrow, with dolichocephaly, high-
down-slanting palpebral fissures, hypertelorism, arched narrow palate, micrognathia, retrognathia,
strabismus, and nystagmus. Developmental and crowded teeth. Respiratory findings include
delay, behavioral problems with expressive diffi- pneumothorax, pulmonary blebs, and, in the worst
culties, and possibly seizures are often found. No cases, emphysema. Cardiac abnormalities include
evidence is found of GH excess, but bone age is aortic root dilatation, aortic regurgitation, ascend-
advanced, and hands and feet are large, with genu ing aortic aneurysm, aortic dissection, mitral
valgum. Rapid growth is most characteristic of regurgitation, mitral valve prolapse, congestive
infancy, and height velocity decreases by mid- heart failure, pulmonary artery dilatation, tricus-
childhood or adolescence, leading in many cases pid valve prolapse, and premature calcification of
to normal adult stature. mitral annulus. The skin reveals striae distensae.
The Weaver syndrome (#277590 WEAVER The disorder results from a mutation in the fibril-
SYNDROME) is associated with prenatal as well lin gene.
88 5 Disorders of Growth

Homocystinuria (*236200 HOMOCYSTINU- elsewhere. The criteria for successful treatment


RIA at 21q22.3) is an autosomal dominant in adults are stringent and are probably the same for
condition with a phenotype somewhat similar the rare cases of gigantism: normal (age/sex-
to that of Marfan syndrome including ectopia adjusted) insulin-like growth factor type 1 (IGF-
lentis. The disorder is owing to mutations in 1), growth hormone (GH) random (GHr) <1 μg/L,
cystathionine β-synthase, which leads to the and a GH nadir (GHn) during oral glucose toler-
laboratory findings of homocystinuria and methi- ance test (OGTT) of <0.4 μg/L.
oninuria. Pectus excavatum or carinatum, osteopo- Precocious puberty of any etiology increases
rosis, biconcave “codfish” vertebrae, kyphoscoliosis, growth in childhood, but early epiphyseal closure
dolichostenomelia, arachnodactyly, and limited and short adult stature will result if untreated (see
joint mobility may be found. Mitral valve prolapse Chap. 9).
is seen as is an increased risk for myocardial infarc- One remarkable case of estrogen-receptor
tion. Developmental delay, psychiatric disorders, defect (#615363 ESTROGEN RESISTANCE;
and seizures are all characteristic of homocystin- ESTRR) in a male led to continued growth into
uria. Treatment involves a low-methionine, cystine- the 25th year, with a bone age delayed so far that
supplemented diet for pyridoxine nonresponders the epiphyses remained open. Lack of estrogen
and pyridoxine supplementation for the roughly effect also led to osteoporosis. This case helped
50 % of affected patients who are classified as establish the understanding that it is estradiol that
pyridoxine responders. advances bone age and stimulates GH secretion.
Syndromes with extra Y chromosomes such A single female case has been identified as well.
as 47XYY or 48XYYY lead to tall stature in Thyrotoxicosis increases height velocity and
childhood and adult life, without any evidence of skeletal age advancement if left untreated (see
increased GH. Klinefelter syndrome (47XXY) Chap. 6).
may be associated with tall stature, but this is not Increased insulin can increase growth rate by
universal (see Chaps. 8 and 9). cross-reaction with IGF-1 receptors. Infants of
diabetic mothers are large at birth, but postnatal
Endocrine Etiologies of Tall Stature growth is normal as insulin effects wane. Fetal
Pituitary gigantism is due to a GH-secreting ade- hyperinsulinism is the stimulus for the excessive
noma in childhood; acromegaly occurs with pitu- fetal growth; neonatal hypoglycemia can be a
itary hypersecretion of GH in an adult. Because serious complication, as the high maternal glu-
the epiphyses are not closed in an affected child, cose concentration that led to the fetal hyperinsu-
height velocity increases, although some of the linism is removed at the time of birth, but the
coarse facial features of an acromegalic appear- insulin secretion continues for days. Children
ance also may be noted. Organomegaly may with β-cell adenomas could have continued rapid
occur, and glucose intolerance or frank diabetes growth during their hyperinsulinemic state, but
may result. Elevated basal GH concentrations their hypoglycemia will most likely be the clini-
and IGF-1 and IGF-BP3 levels confirm the diag- cal condition that brings them to diagnosis and
nosis. A pathologic rise in growth hormone after treatment first (see Chap. 12).
glucose administration or after TRF administra- Beckwith–Wiedemann syndrome (#130650
tion is another feature of somatotropic adenomas. BECKWITH-WIEDEMANN SYNDROME;
McCune–Albright syndrome may have, as one BWS imprinting at 11p15.5) presents with large
feature, pituitary gigantism due to autonomous newborn length and weight, omphalocele (exom-
GH secretion (see Chap. 9). Likewise, multiple phalos), and macroglossia. Body size remains
endocrine neoplasia 1 (MEN 1) may include large throughout childhood, with an advanced
excessive GH secretion, and if, as rarely occurs, bone age. Life-threatening hypoglycemia occurs
it occurs before epiphyseal fusion, pituitary because of hyperinsulinism due to pancreatic
gigantism will result (see Chap. 10). Treatment is hyperplasia. In addition, hemihypertrophy,
accomplished by somatostatin analogues, radia- diastasis recti, hepatomegaly, and cardiomegaly
tion therapy, or surgery, all of which are discussed are seen. A large fontanelle with a prominent
Suggested Readings 89

occiput, a metopic ridge, coarse facial features, 2. Cousminer DL, Berry DJ, Timpson NJ, Ang W,
Thiering E, Byrne EM, Taal HR, Huikari V, Bradfield
and linear ear lobe creases with posterior helical
JP, Kerkhof M, Groen-Blokhuis MM, Kreiner-Moller
indentations occur. In addition, fetal adrenocortical E, Marinelli M, Holst C, Leinonen JT, Perry JR,
cytomegaly and large kidneys with medullary Surakka I, Pietilainen O, Kettunen J, Anttila V,
dysplasia are found. Cryptorchidism is described, Kaakinen M, Sovio U, Pouta A, Das S, Lagou V,
Power C, Prokopenko I, Evans DM, Kemp JP, St
with a tendency to neoplasia including Wilms
Pourcain B, Ring S, Palotie A, Kajantie E, Osmond C,
tumor, hepatoblastoma, adrenal carcinoma, and Lehtimaki T, Viikari JS, Kahonen M, Warrington NM,
gonadoblastoma. Beckwith–Wiedemann syn- Lye SJ, Palmer LJ, Tiesler CM, Flexeder C,
drome results from overexpression of the pater- Montgomery GW, Medland SE, Hofman A,
Hakonarson H, Guxens M, Bartels M, Salomaa V,
nally expressed genes or a lack of expression of the
ReproGen C, Murabito JM, Kaprio J, Sorensen TI,
maternally expressed genes within the 11p15 Ballester F, Bisgaard H, Boomsma DI, Koppelman
region. Wilms tumor is associated with uniparental GH, Grant SF, Jaddoe VW, Martin NG, Heinrich J,
disomies of paternal origin and gain of methylation Pennell CE, Raitakari OT, Eriksson JG, Smith GD,
Hypponen E, Jarvelin MR, McCarthy MI, Ripatti S,
at ICR1 (the opposite of hypomethylation at ICR1
Widen E, Early Growth Genetics Consortium.
found in the Russell–Silver dwarfism above). Genome-wide association and longitudinal analyses
reveal genetic loci linking pubertal height growth,
Diagnosis and Treatment of Tall pubertal timing and childhood adiposity. Hum Mol
Genet. 2013;22(13):2735–47.
Stature 3. Munoz-Hoyos A, Molina-Carballo A, Augustin-
Generally, the historical and physical manifesta- Morales M, Contreras-Chova F, Naranjo-Gomez A,
tions of the conditions listed earlier are evident Justicia-Martinez F, Uberos J. Psychosocial dwarfism:
during the history and physical examination. psychopathological aspects and putative neuroendo-
crine markers. Psychiatry Res. 2011;188(1):96–101.
Pituitary gigantism is associated with an elevated
4. Ahmad T, Garcia-Filion P, Borchert M, Kaufman F,
IGF-1 concentration but only if precocious Burkett L, Geffner M. Endocrinological and auxo-
puberty is ruled out since during precocious logical abnormalities in young children with optic
puberty, IGF-1 concentrations increase higher nerve hypoplasia: a prospective study. J Pediatr.
2006;148(1):78–84.
than in age-matched controls and may be con-
5. Chernausek SD, Backeljauw PF, Frane J, Kuntze J,
fused with acromegalic values. Basal GH con- Underwood LE. Long-term treatment with recombi-
centration is usually elevated in pituitary nant insulin-like growth factor (IGF)-I in children
gigantism when compared with the low concen- with severe IGF-I deficiency due to growth hormone
insensitivity. J Clin Endocrinol Metab. 2007;92(3):
trations seen in normal subjects (a rare situation
902–10.
in which a single GH determination may lead to 6. Clayton PE, Cianfarani S, Czernichow P, Johannsson
a diagnosis). Pituitary gigantism also responds G, Rapaport R, Rogol A. Management of the child
with GH secretion to agents that normally do not born small for gestational age through to adulthood: a
consensus statement of the International Societies of
stimulate GH secretion, agents such as glucose
Pediatric Endocrinology and the Growth Hormone
(normally glucose suppresses GH), gonadotropin- Research Society. J Clin Endocrinol Metab. 2007;
releasing hormone (GnRH), and thyrotropin- 92(3):804–10.
releasing factor (TRF). Pituitary gigantism may 7. Cohen P, Rogol AD, Deal CL, Saenger P, Reiter EO,
Ross JL, Chernausek SD, Savage MO, Wit
be treated with transsphenoidal microadenomec-
JM. Consensus statement on the diagnosis and treatment
tomy if the tumor is approached early. of children with idiopathic short stature: a summary of
the Growth Hormone Research Society, the Lawson
Wilkins Pediatric Endocrine Society, and the European
Society for Paediatric Endocrinology Workshop. J Clin
Suggested Readings Endocrinol Metab. 2008;93(11):4210–7.
8. David A, Hwa V, Metherell LA, Netchine I,
1. Growth Hormone Research Society. Consensus Camacho-Hubner C, Clark AJ, Rosenfeld RG,
guidelines for the diagnosis and treatment of growth Savage MO. Evidence for a continuum of genetic,
hormone (GH) deficiency in childhood and adoles- phenotypic, and biochemical abnormalities in chil-
cence: summary statement of the GH Research dren with growth hormone insensitivity. Endocr Rev.
Society. GH Research Society. J Clin Endocrinol 2011;32(4):472–97.
Metab. 2000;85(11):3990–3.
90 5 Disorders of Growth

9. Keil MF, Stratakis CA. Pituitary tumors in childhood: 27. Poidvin A, Touze E, Ecosse E, Landier F, Bejot Y, Giroud
update of diagnosis, treatment and molecular genet- M, Rothwell PM, Carel JC, Coste J. Growth hormone
ics. Expert Rev Neurother. 2008;8(4):563–74. treatment for childhood short stature and risk of stroke in
10. Wit JM. Definition and subcategorization of idio- early adulthood. Neurology. 2014;83(9):780–6.
pathic short stature: between consensus and contro- 28. Savage MO. Insulin-like growth factors, nutrition and
versy. Horm Res Paediatr. 2011;76 Suppl 3:3–6. growth. World Rev Nutr Diet. 2013;106:52–9.
11. Mauras N. Strategies for maximizing growth in 29. Fine RN, Martz K, Stablein D. What have 20 years of
puberty in children with short stature. Pediatr Clin data from the North American Pediatric Renal
North Am. 2011;58(5):1167–79. Transplant Cooperative Study taught us about growth
12. Collett-Solberg PF. Update in growth hormone therapy following renal transplantation in infants, children,
of children. J Clin Endocrinol Metab. 2011;96(3): and adolescents with end-stage renal disease? Pediatr
573–9. Nephrol. 2010;25(4):739–46.
13. Deodati A, Cianfarani S. Impact of growth hormone 30. Ulijaszek SJ. The international growth standard for
therapy on adult height of children with idiopathic short children and adolescents project: environmental influ-
stature: systematic review. BMJ. 2011;342:c7157. ences on preadolescent and adolescent growth in
14. Bondy CA, Turner Syndrome Study Group. Care of weight and height. Food Nutr Bull. 2006;27(4 Suppl
girls and women with Turner syndrome: a guideline Growth Standard):S279–94.
of the Turner Syndrome Study Group. J Clin 31. Kjaer I. Mechanism of human tooth eruption: review
Endocrinol Metab. 2007;92(1):10–25. article including a new theory for future studies on the
15. Divall SA, Radovick S. Growth hormone and treat- eruption process. Scientifica. 2014;2014:341905.
ment controversy; long term safety of rGH. Curr 32. Styne DM. Growth. In: Greenspan FS, Strewler FJ,
Pediatr Rep. 2013;1(2):128–32. editors. Greenspan’s basic and clinical endocrinology.
16. Deal CL, Tony M, Hoybye C, Allen DB, Tauber M, 9th ed. Stamford, CT: Appleton and Lange; 2011.
Christiansen JS. Growth Hormone Research Society 33. Cooke DW, Divall SA, Radovick S. Normal and aber-
workshop summary: consensus guidelines for recombi- rant growth. In: Melmed S, Polonsky KS, Larsen PR,
nant human growth hormone therapy in Prader-Willi syn- Kronenberg HM, editors. Williams textbook of endo-
drome. J Clin Endocrinol Metab. 2013;98(6):E1072–87. crinology. 12th ed. Philadelphia, PA: Saunders
17. Rivkees SA, Hager K, Hosono S, Wise A, Li P, Rinder Elsevier; 2015.
HM, Gruen JR. A highly sensitive, high-throughput 34. Greulich WW. Radiographic atlas of skeletal develop-
assay for the detection of Turner syndrome. J Clin ment of the hand and wrist. Stanford, CA: Stanford
Endocrinol Metab. 2011;96(3):699–705. University Press; 1959 [repr. 1970].
18. Backeljauw PF, Dattani MT, Cohen P, Rosenfeld 35. Rogol AD. Treatment of growth hormone deficiency
RG. Chapter 10—Disorders of growth hormone/ in children Upto Date 2015 http://www.uptodate.com/
insulin-like growth factor secretion and action. In: contents/treatment-of-growth-hormone-deficiency
Sperling MA, editor. Pediatric endocrinology. 4th ed. in-children.
Philadelphia, PA: Elsevier; 2014. p. 291–404. 36. Tanner JM, Davies PS. Clinical longitudinal standards
19. Wit JM, Oostdijk W, Losekoot M. Spectrum of for height and height velocity for North American
insulin-like growth factor deficiency. Endocr Dev. children. J Pediatr. 1985;107(3):317–29.
2012;23:30–41. 37. Kelly A, Winer KK, Kalkwarf H, Oberfield SE, Lappe
20. Wit JM, Hero M, Nunez SB. Aromatase inhibitors in J, Gilsanz V, Zemel BS. Age-based reference ranges
pediatrics. Nat Rev Endocrinol. 2012;8(3):135–47. for annual height velocity in US children. J Clin
21. Wolfgram PM, Carrel AL, Allen DB. Long-term Endocrinol Metab. 2014;99(6):2104–12.
effects of recombinant human growth hormone 38. Rogol AD. Diagnostic approach to children and
therapy in children with Prader-Willi syndrome. Curr adolescents with short stature and-adolescents-with-
Opin Pediatr. 2013;25(4):509–14. short-stature. http://www.uptodate.com/contents/
22. Horton WA, Hall JG, Hecht JT. Achondroplasia. diagnostic-approach-to-children-and-adolescents-
Lancet. 2007;370(9582):162–72. with-short-stature.
23. Ranke MB. Treatment of children and adolescents 39. Rogol AD, Richmond AJ. The child with tall stature
with idiopathic short stature. Nat Rev Endocrinol. and/or abnormally rapid growth. http://www.upto-
2013;9(6):325–34. date.com/contents/the-child-with-tall-stature-and-or-
24. Rogol AD, Hayden GF. Etiologies and early diagnosis abnormally-rapid-growth.
of short stature and growth failure in children and 40. Rogol AD. Treatment of growth hormone deficiency
adolescents. J Pediatr. 2014;164(5 Suppl):S1–14 e16. in children. http://www.uptodate.com/contents/
25. Eugster EA, Pescovitz OH. Gigantism. J Clin treatment-of-growth-hormone-deficiency-in-children.
Endocrinol Metab. 1999;84(12):4379–84. 41. Rogol AD, Richmond AJ. Diagnosis of growth hor-
26. Brooks AJ, Waters MJ. The growth hormone receptor: mone deficiency in children. http://www.uptodate.
mechanism of activation and clinical implications. com/contents/diagnosis-of-growth-hormone-deficiency-
Nat Rev Endocrinol. 2010;6(9):515–25. in-children.
Disorders of the Thyroid Gland
6

The hormones of the thyroid gland exert impor- (primary hypothyroidism), TSH increases. TSH is
tant effects on growth and development including released in a pulsatile manner with increased
early effects on the development of the central release at night. Secondary hypothyroidism (inad-
nervous system (CNS), regulation of tempera- equate or absence of pituitary TSH) and tertiary
ture, and influences on metabolism. Disorders of hypothyroidism (inadequate or absence of hypo-
the thyroid gland are common forms of endocrine thalamic TRF) are differentiated from primary
disease in childhood, and the primary care pro- hypothyroidism by the finding of normal or
vider will definitely encounter some of these. decreased serum TSH; in the secondary form,
serum TSH is nondetectable before or after TRF
administration; in the tertiary form, TSH is measur-
Normal Thyroid Physiology able and increases from low levels to an abnor-
and Anatomy mally prolonged elevation after TRF although even
normal responses are reported. In the presence of
The production of thyroid hormones by the thyroid autonomous and excessive production of endoge-
gland is regulated by the hypothalamus and the nous thyroid hormone (as found in Graves’ dis-
pituitary gland (Fig. 6.1). Hypothalamic thyrotropin- ease) or administration of exogenous thyroid
releasing hormone (thyrotropin-releasing factor or hormone, the release of TSH is suppressed to non-
TRF in some publications), a three-amino-acid measurable levels in sensitive TSH assays.
peptide, is released from the median eminence into Thyroid hormones are composed of two tyro-
the pituitary portal circulation. Although TRF has sine molecules, an α(inner)- and a β(outer)-ring,
many functions in the CNS, for our purposes, it and a variable amount of iodine. The inner or
stimulates the pituitary gland to release thyroid- alpha ring is closest to the alanine side chain, and
stimulating hormone (TSH) into the general circu- the outer or beta ring is farthest away. They are
lation, where it reaches the thyroid gland and synthesized in the following sequential steps:
stimulates the production and release of endoge-
nous thyroid hormones. Thyroid hormones exert 1. Active transport of plasma iodide into the thy-
feedback inhibition on the hypothalamic–pituitary roid follicular cell by the iodine/sodium sym-
axis and suppress TSH secretion to reach an equi- porter (or the iodine pump) stimulated by
librium of euthyroidism. Thyroxine rather than T3 TSH; thyroglobulin is found in the colloid of
appears to be most important in suppression of the thyroid gland, and this is the site of organi-
TSH and feedback inhibition. In the absence of fication of the tyrosine residues attached to
adequate thyroid hormone production and release thyroid globulin.

© Springer International Publishing Switzerland 2016 91


D.M. Styne, Pediatric Endocrinology, DOI 10.1007/978-3-319-18371-8_6
92 6 Disorders of the Thyroid Gland

a Third ventricle Hypothalamus

Neuroendocrine
cell nuclei TRF secreting cells

Superior
hypophyseal Stalk
artery

Long portal
vessels Inferior
hypophyseal
artery
Thryrotrophs

Anterior Posterior
pituitary pituitary

Short portal
Hormone vessel
secretion

TSH

T4 and T3

Fig. 6.1 The hypothalamic–pituitary–thyroid axis. (a) would demonstrate secondary hypothyroidism), insuf-
Normally a balance exists between thyroid hormones ficient TSH stimulates the thyroid gland to produce
and thyroid-stimulating hormone (TSH) due to a func- adequate thyroid hormone. (d) In hyperthyroidism due
tioning feedback loop. (b) In primary hypothyroidism, to Graves’ disease, the autonomous production of thy-
insufficient thyroid hormones are produced to suppress roid hormone by thyroid-stimulating immunoglobulin
TSH to normal. (c) In secondary or tertiary hypothy- (TSI) and TSH receptor antibody suppresses TSH to
roidism (tertiary hypothyroidism is demonstrated in nondetectable levels
this figure; a red X put over the anterior pituitary gland

2. Organification of the tyrosine molecules is or tetraiodothyronine or T4) or one DIT with


accomplished by the addition of one iodine at one MIT to produce 3,5,3′-triiodothyronine
the 3 position [causing the formation of (triiodothyronine or T3).
monoiodotyrosine (MIT)] or two iodines at 3. The release of T4 and T3 from the thyroid gland
the 3 and 5 positions [forming diiodotyrosine is stimulated by TSH which interacts with the
(DIT)] by thyroid peroxidase at the luminal G protein seven-transmembrane domain cellu-
cell membrane stimulated by TSH. Mature lar membrane receptor causing stimulation of
thyroid hormones are composed of two iodin- adenyl cyclase to produce cyclic AMP ulti-
ated tyrosine molecules with an alanine side mately causing the protein-mediated release of
chain: the coupling of one DIT with one DIT thyroid hormones from thyroglobulin and
to form 3,5,3′,5′-tetraiodothyronine (thyroxine release of the hormones into the circulation.
Normal Thyroid Physiology and Anatomy 93

b Third ventricle Hypothalamus

Neuroendocrine
cell nuclei TRF secreting cells

Superior
hypophyseal Stalk
artery

Long portal
vessels Inferior
hypophyseal
artery
Thryrotrophs

Anterior Posterior
pituitary pituitary

Short portal
Hormone vessel
secretion

TSH

T4 and T3

Fig. 6.1 (continued)

4. TSH stimulates entry of thyroglobulin into the Circulating T4 is bound predominantly to


follicular cell by pleocytosis, where it is serum proteins; 75 % is bound to thyroid-binding
metabolized and iodine is subsequently globulin (TBG), 20 % to transthyretin (thyroxine-
released and reused in the process again. binding prealbumin or TBPA), and 5 % to albu-
min, with only 0.02–0.06 % present in the
Although both T4 and T3 are released from the circulation as free thyroxine (FT4); about 0.3 %
thyroid gland, most of the circulating T3 is of T3 circulates as free T3 (FT3). Free T3 is the
derived from peripheral deiodination of the metabolically active form of thyroid hormone,
β-ring of T4, except in the case of hyperthyroid- whereas protein-bound T3 and T4 may be consid-
ism, in which excessive T3 secretion from the ered reservoirs of hormone in equilibrium with
thyroid gland may occur. T3 is the metabolically the metabolically active free hormone. The thy-
active form of thyroid hormone. Reverse T3 (RT3) roid binding proteins are affected by genetic con-
or 3,3′,5′-triiodothyronine is a metabolically ditions, are decreased by androgens, and are
inert product of the peripheral deiodination of the raised by estrogens, thereby changing the total T4
α-ring of T4; this form is found in greater amounts measurement. Thus, free T4 is the preferable
in the fetus or during severe illness. measurement in an accurate assay; there are older
94 6 Disorders of the Thyroid Gland

c Third ventricle Hypothalamus

Neuroendocrine
cell nuclei TRF secreting cells

Superior
hypophyseal Stalk
artery

Long portal
vessels Inferior
hypophyseal
artery
Thryrotrophs

Anterior Posterior
pituitary pituitary

Short portal
Hormone vessel
secretion

TSH

T4 and T3

Fig. 6.1 (continued)

methods such as resin T3 uptake (RT3U) to boxylate transporter 8 gene (MCT8) (#300523
account for variations in protein binding after ICD+ALLAN-HERNDON-DUDLEY
measuring total T4 but they are rarely used now. SYNDROME; AHDS) and is characterized by
T3 enters the cell through thyroid hormone low serum-free thyroxine, low serum rT3, but
transporters located in the cell membrane. There increased serum triiodothyronine (T3) and normal
are several types of thyroid hormone transport- or mildly increased thyroid-stimulating hormone
ers including Na+/taurocholate cotransport- (TSH) along with dysmorphology and profound
ing polypeptide, the Na+/independent organic mental and muscular developmental delay.
anion transporting polypeptide (OATP) family Free T3 binds to nuclear thyroid hormone pro-
operating mainly in the central nervous system, tein receptors (TR) that modulate thyroid
the heterodimeric L-type amino acid transport- hormone-dependent gene expression. There are
ers (LAT1, LAT2), and the monocarboxylate two forms of receptors, TRalpha and TRbeta
transporter (MCT) family which is widespread with two forms of each receptor; the four result-
in various organs. The Allan–Herndon–Dudley ing receptors are expressed in different thyroid
syndrome is due to mutations in the monocar- responsive tissues in the body. Thyroid hormone
Fetal Thyroid Development in the Newborn 95

d Third ventricle Hypothalamus

Neuroendocrine
cell nuclei TRF secreting cells

Superior
hypophyseal Stalk
artery

Long portal
vessels Inferior
hypophyseal
artery
Thryrotrophs

Anterior Posterior
pituitary pituitary

Short portal
Hormone vessel
secretion

TSH

TSI
TSHR AB

T4 and T3

Fig. 6.1 (continued)

receptors are similar to steroid receptors in Once the DNA/TR/thyroid hormone/coactivator


structure and function and are members of the complex is formed, RNA polymerase transcribes
steroid hormone superfamily. The receptors are DNA into mRNA which leads to translation of
present either as monomers (TR), heterodimers the mRNA on ribosomes and the production of
with retinoid X receptor (TR/RXR), or homodi- proteins or peptides predicted by the hormone
mers (TR/TR). TR/RXR heterodimers are the that result in changes in cell function.
most transcriptionally active complex. In the
absence of hormone, TR exists in a complex with
corepressor proteins binding to hormone response Fetal Thyroid Development
elements (HREs) in DNA in an inactive state. in the Newborn
Binding of thyroid hormone results in a confor-
mational change in TR which displaces corepres- The thyroid gland originates from the embryonic
sors from the receptor/DNA complex while pharyngeal floor between the first and second
adding coactivator proteins to the complex. pharyngo-bronchial pouch near a site later identi-
96 6 Disorders of the Thyroid Gland

fied as the foramen cecum, noted after birth as a to the production of T3 the active form of thyroid
pit at the back of the tongue. The normal thyroid hormone. In the fetus, the majority of T4 under-
gland descends to the customary location at the goes deiodination of the inner ring, leading to the
thyroid cartilage anterior to the trachea. In some formation of RT3 (compared with active T3) in
cases of congenital hypothyroidism, the thyroid parallel with concentrations of T4 lower than
gland fails to descend far enough (sublingual or found in postnatal life. Late in gestation, outer-
lingual thyroid is the result) or descends too far, ring deiodination of thyroxine increases, forming
sometimes ending up as far inferior as the medi- more T3, and serum T3 finally begins to increase
astinum. An ectopic thyroid gland may be too as well. At term, fetal TSH and T4 are slightly
small or dysplastic to produce adequate thyroid higher than those found in normal adult concen-
hormone, resulting in congenital hypothyroid- trations; fetal serum RT3 is markedly higher than
ism. A thyroid gland in the normal location also in the adult, and fetal serum T3 is considerably
may be small (thyroid dysplasia or hypoplasia), lower. Thus, the fetus is normally in a relatively
or, in the most severe case, absent in athyreosis. decreased state of thyroid action compared with
One lobe may be absent, or an extra lobe, the the postnatal state.
pyramidal lobe, may develop out of an embry- Serum values of TBG and other thyroid-
onic remnant of the thyroglossal duct cyst, lead- binding proteins increase with advancing gesta-
ing to asymmetry in appearance but possibly tion, as do other serum proteins.
normal function through compensatory hypertro- Serum concentrations of the thyroid hor-
phy. Thyroid dysgenesis may occur due to abnor- mones found in the fetus reflect maturation of
malities in the homeobox genes (PAX8, TTF1, the fetal thyroid gland, as little of the maternal
FOXE1 and NKX2-5, which are responsible for thyroid hormones and virtually none of the
thyroid embryogenesis) in the approximately maternal TSH cross the placenta. During preg-
2 % of cases that are familial; however, most nancy hCG will tend to stimulate thyroid func-
cases of thyroid dysgenesis occur sporadically. tion, and estrogens will likewise raise protein
A thyroglossal duct cyst connects the foramen binding levels increasing total thyroxine.
cecum to the final location of the thyroid gland However, it is estimated that as many as 15 % of
and may be present after fetal life, sometimes women have subclinical hypothyroidism during
containing functional thyroid tissue. If a thyro- pregnancy. Subclinical hypothyroidism in the
glossal duct cyst is removed without insuring the mother can lead to spontaneous abortion, pre-
presence of other thyroid tissue, the child may be eclampsia, gestational hypertension, gestational
rendered hypothyroid by the surgery. diabetes, and preterm delivery. A mother on thy-
By 10–11 weeks of gestation, the human thy- roxine therapy is estimated to require a 50 %
roid gland shows follicular organization and higher dose during pregnancy. There is evidence,
demonstrates the ability to concentrate iodine. By although it is not found in all studies, that a
this time, TRF is demonstrable in the fetal hypo- mother with inadequately or untreated hypothy-
thalamus, and TSH is found in the fetal pituitary roidism will have an offspring with slightly
and circulation. The pituitary–hypothalamic por- decreased IQ, suggesting the importance of the
tal system is developing around this time and is small amount of T4 that does cross the placenta
mostly mature by 20 weeks’ gestation. Most of before the fetal thyroid gland matures.
the components of thyroid physiology are in Hypothyroidism in both mother and fetus may
place by the second half of gestation. Thus, TSH lead to severe developmental delay. If the mother
stimulation of thyroid function begins approxi- produces a normal amount of thyroxine, even if
mately midgestation. the fetus is profoundly hypothyroid, early treat-
Monodeiodination of the inner ring α of ment after birth will usually allow normal intel-
thyroxine leads to reverse T3 (RT3), an inactive lectual development demonstrating the
moiety (compared with active T3), and monode- importance of the maternal contribution to thy-
iodination of the outer ring β of thyroxine leads roid physiology.
Laboratory Evaluation 97

After delivery, serum TSH quickly increases Serum thyroglobulin (TBG) assays are
and peaks at 30 min; T4 is secreted in response invoked to determine the presence of thyroid tis-
and increases by 24–72 h to values equivalent to sue as only this organ produces thyroglobulin.
those found in adult hyperthyroidism, with a slow Endogenous Antibodies to TBG however affect
decrease toward adult normal values over the fol- results of TBG determinations.
lowing weeks, months, and years. Serum T3 Ultrasensitive TSH assays are now widely
exhibits a primary increase in the hours after birth available to measure TSH accurately to 0.5 mIU/
after the umbilical cord is cut because of increased mL or less. TSH values are dependent upon assay
outer-ring deiodination of thyroxine and exhibits characteristics, so it is best to use the same assay
a secondary increase at 24–72 h due to the increase sequentially when following a patient over time,
in T4 secretion; RT3 decreases after birth because often difficult when the laboratory you are depen-
of the switch from inner-ring deiodination to dent upon in a medical system switches to a
outer-ring deiodination; TBG remains relatively cheaper contracted send-out laboratory or uses a
constant after birth. Localized deiodination in new assay platform. TSH values also show a
brown adipose tissue which is present in the neo- diurnal rhythm of peaks at midnight and nadirs at
nate but not in any quantity in the adult leads to noon in most situations. Values vary between
localized production of T3 and is felt to enhance individuals more than values repeatedly taken
thermogenesis in the newborn after birth. from a single individual, so the best comparisons
are in the same assay in the same individual if
treatment is invoked. Lastly, the determination of
Laboratory Evaluation the upper limit of normal TSH values in children
may not always be straightforward. Longitudinal
In the past, total serum T4 was the most common study of children with only slightly elevated TSH
measurement available. However, the total T4 values up to 7.5 mIU/L was followed with a
value must be adjusted for the effects of available descent into the normal range or stability with no
serum proteins on total T4 measurement. If thy- evidence of disease in most demonstrating that
roid-binding proteins are altered by genetic con- normal values for TSH may be higher in children
ditions, the total T4 value will change with no than adults. Further, values only slightly below
change in the free moiety and without biological the normal reference interval do not necessarily
complications. One older method to determine indicate hyperthyroidism. As with all laboratory
thyroid-binding proteins invokes the use of a tests, clinical correlation is necessary in interpret-
resin T3 (RT3U) assay which serves as a surro- ing this test.
gate test for the direct measurement of the protein A patient with Graves’ disease will have a
effects on total thyroid hormone levels. The total value of serum TSH below the normal reference
T4 is multiplied by the RT3U thereby calculating range (often less than 0.5 mIU/mL) because of
the free thyroxine index. This complex procedure suppression by the autonomous secretion of thy-
is now usually replaced by the direct measure- roid hormones. A patient with secondary (pitu-
ment of FT4 by RIA or more accurately by equi- itary defect lacking TSH production) or tertiary
librium dialysis methods, although this later (hypothalamic defect due to lack of TRF produc-
method requires more effort, is more expensive, tion) hypothyroidism will have variable results,
and is generally available in national laborato- whereas a normal adult patient will have a value
ries. Labeled analog or labeled antibody-free T4 between 0.3 and 3.04 mIU/L (check the labora-
assays are more readily available. As always, tory used for their exact standards for age, or
technical issues can affect results, so clinical cor- refer to Chap. 15). In primary hypothyroidism or
relation or correlation with TSH values is help- compensated hypothyroidism, serum TSH is
ful. Thyroid hormone values must be interpreted elevated. The combination of serum FT4 and
by age in childhood, so choice of laboratories TSH determination will allow the differential
with pediatric standards is essential. diagnosis of primary from secondary or tertiary
98 6 Disorders of the Thyroid Gland

hypothyroidism. Differentiation between sec- more strongly suggesting a malignancy, surgical


ondary hypothyroidism and tertiary hypothy- excision biopsy is a better approach.
roidism is more difficult and may require a Radioiodine imaging of a thyroid gland may
dynamic TRF test. A dose of 200 mg of TRF is be used in the differential diagnosis of nodules of
given intravenously (i.v.) and TSH measured at the thyroid gland to determine whether they are
0, 10, 15, 30, 60, 90, 120, and 180 min (or at functional or “hot” compared with nonfunctional
several of these times in some protocols). A nor- or “cold” nodules. Imaging may be used for the
mal response is an increase in TSH to at least localization of an ectopic thyroid gland. However,
10 mIU/mL after about 15 min; in secondary in a clear case of Hashimoto thyroiditis with clas-
hypothyroidism, no increase is seen in TSH, and sic history, findings, and laboratory findings, lit-
in tertiary hypothyroidism, the increase may be tle is added by a thyroid scan. Radioiodine or
delayed until 60–120 min, and the TSH may technetium uptake will be used in some cases in
continue to increase during the 180-min period. the diagnosis of Graves’ disease or in preparation
The TRF test also may be used in the diagnosis for radioiodine therapy for Graves’ disease or
of hyperthyroidism, as no increase in TSH will thyroid carcinoma.
occur after TSH with the autonomous thyroxine The scanning of a neonate or infant presents
secretion characteristic of Graves’ disease. The particular problems; the radioiodine tracer, which
TRF test is rarely invoked in Graves’ disease or is administered orally or by nasogastric (NG)
other conditions with the development of newer tube, may be spit up by the infant and may leave
laboratory techniques including ultrasensitive tracts of radioactivity on the skin, falsely suggest-
TSH and newer thyroid-stimulating antibody ing thyroid tissue on imaging. Injectable perchlo-
tests. rate is an alternative tracer which can be given IV.
Assays for T3 or free T3 are of interest in the It is recommended that the child be fed to elimi-
evaluation of hyperthyroidism but not usually nate tracer uptake in salivary glands and to foster
necessary in the diagnosis of hypothyroidism. the onset of sleep during the scan. Further, a neo-
Measurement of reverse T3 (RT3) is rarely invoked nate suspected of having hypothyroidism may
except in some cases of nonthyroidal illness. have no detectable thyroid gland on scan just
after birth, but after months pass, a normal gland
may be detected; this may be due to the transient
Imaging of Thyroid presence of thyroid-binding inhibitory immuno-
globulin in the neonatal circulation that was
Ultrasonographic scanning of the thyroid gland is passed from the mother (if the mother has symp-
useful to determine the location and size of a thy- tomatic or asymptomatic autoimmune thyroid
roid gland mass; however, it is not as reliable in disease) to the fetus. Application of iodine-con-
finding ectopic glands in the diagnosis of con- taining cleaning compounds to the skin of the
genital hypothyroidism. A scan may differentiate neonate may cause transient hypothyroidism by
cystic from solid masses and the relation of the inhibiting neonatal thyroid function. A congeni-
mass to neck structures. If a solitary nodule is tal iodine-trapping defect (#274400THYROID
solid, it is more likely to be carcinoma than if it is DYSHORMONOGENESIS 1; TDH1) will elim-
a simple cyst. Ultrasonography also can be used inate thyroidal uptake of radioiodine and suggest
to guide a biopsy needle for fine-needle aspira- aplasia but appear the same as these transient
tions. However, if a needle biopsy is obtained, conditions on US and scan. The demonstration of
the pathology service must have experience in an ectopic or hypoplastic gland in a neonate on
accurately interpreting the results. Some suggest an acceptable-quality scan, however, is good evi-
that because excision may be indicated in the dence that the child has a permanent defect in thy-
case of single cold nodules in any case, excision roid function. In the differential diagnosis of
biopsy should be performed for both diagnosis congenital goiter, perchlorate discharge test may
and treatment of suspected cancer. With features be used in which radioactive thyroid uptake is
Disorders of the Thyroid Gland 99

determined and then determined a second time focus on the detection of soft tissue anterior to the
after administration of potassium perchlorate, tracheal cartilage. The size of the thyroid gland
which causes the discharge of nonorganified can be estimated as 5 g of thyroid tissue is the
iodine; if the turnover of iodine is much higher equivalent of a teaspoon size of tissue and 15 g of
with perchlorate than without, an autosomal a tablespoon. The thyroid gland increases in size
recessive enzyme deficiency causing an organifi- with growth until the teenager reaches a gland
cation defect is present. size of about 10 g. The margins of the goiter
An important consideration when performing should be noted distinctly from the neighboring
radioactive thyroid-uptake determinations for sternocleidomastoid muscle, and measurement
hyperthyroidism is to measure the uptake early, should be recorded. The longest diagonal length
such as at 4 h, as well as at 24 h, because a quick of each lobe, the shortest diagonal length of each
turnover of radioactive iodine, as occurs in lobe, and the height of the isthmus may be
Graves’ disease, may lead to a high uptake at 4 h recorded for comparison at future visits to deter-
but a low uptake at 24 h, so that a sole late scan mine the effect of treatment. Some observers find
may falsely eliminate hyperthyroidism as a diag- it useful to trace the outline of the gland on a
nosis. Scanning can be done with 123I or techne- piece of tissue paper and to include it in the
tium 99 pertechnetate; the former must be used patient’s record for future comparison. An esti-
before thyroxine is administered (if indicated), mation of the percentage the gland appears to be
whereas the latter may be used even if thyroxine increased over normal size may be accomplished
has already been administered. The ingestion of (i.e., a slightly or moderatedly enlarged gland
excessive iodine or the use of contrast agents will might be 25–50 % increased, whereas an obvi-
decrease the ability to perform a scan with 123I for ously enlarged gland may be 100 % increased).
a considerable period; in fact the iodine status of While a homogeneous goiter most likely is due to
the individual if outside of the normal intake Hashimoto’s thyroiditis in a child or teenager,
range can affect the results of the test and iodine confirmatory tests must be performed.
intake is increasing with many foods such as Ultrasonography may detect fetal goiters. This
bread and salt augmented with iodine. A low could be due to exogenous agents given to the
iodide diet is characteristically instituted before mother or a metabolic form of hypothyroidism in
radioactive iodine treatment for thyroid carci- the fetus. There are some reported cases of intra-
noma to ensure maximal uptake. uterine treatment of such patients with thyroid hor-
mone, but this is not presently standard of care.
The thyroid functional status is next determined
Disorders of the Thyroid Gland in a patient with a goiter. Most goiters in the neo-
nate will be diagnosed by means of the TSH value
Goiter on the newborn screening programs and will be
due to maternal ingestion of a goitrogen [such as
An enlargement of the thyroid gland is a goiter; methimazole or propylthiouracil (PTU) for treat-
the affected patient may be euthyroid, hypothy- ment of maternal hyperthyroidism] or an inborn
roid, or hyperthyroid, so the finding of goiter does error of thyroid biosynthesis. The majority of goi-
not indicate the diagnosis (Table 6.1). A goiter ters in the late childhood through adolescent years
may be large enough to be visible to the patient or will be due to euthyroid or hypothyroid Hashimoto
parent or may be subtle enough to be noted only thyroiditis, followed by a lower incidence of
on directed physical examination. If the patient hyperthyroidism due to Graves’ disease. A minor-
swallows water during the examination, the ity of indeterminate goiters of mild degree may
mobile nature of the thyroid gland should iden- persist for years; these may be colloid goiters, a
tify it as it rises and falls on swallowing. diagnosis that is established only after all other
Examination of the neonate or infant should diagnoses are eliminated. Colloid goiter occurs
100

Table 6.1 Etiologies of thyroid enlargement


Disorder Goiter Nodule Free T4 TSH Other features Inheritance
Autoimmune Usually Usually not Usually low but Usually high but Positive antithyroid peroxidase or Autosomal dominant, recessive,
hypothyroidism could be Nl or could be Nl or antithyroglobulin antibodies or sporadic; may be part of
(Hashimoto thyroiditis) high in euthyroid low in euthyroid autoimmune
Hashimoto Hashimoto polyendocrinopathy syndromes
Acute or suppurative One lobe or overall No High transiently Low transiently Painful, may originate through
thyroiditis enlargement at beginning at beginning pyriform sinus, treated with
antibiotics
Subacute thyroiditis Usually No High transiently Low transiently Painful, may be treated with
at beginning at beginning anti-inflammatory agents for pain
but since differentiation between
bacterial and viral infection may be
difficult, antibiotics may be used
Adenoma Yes High if functional Low if functional
or Nl if not or Nl if not
Papillary carcinoma Yes Usually normal Usually normal History of radiation? Sporadic or autosomal
dominant
Follicular carcinoma Yes Usually normal Usually normal History of radiation? Sporadic or autosomal
dominant
6

Medullary carcinoma Yes Normal Normal Calcitonin high in basal, Sporadic or familial medullary
pentagastrin-, or calcium- carcinoma of the thyroid
stimulated state; ret mutation (FMCT) and may be associated
positive even before calcitonin with MEN 2 or MEN 3
increases
FMCT familial medullary carcinoma of the thyroid, MEN multiple endocrine neoplasia
Disorders of the Thyroid Gland
Hypothyroidism 101

mainly in areas deficient in adequate iodide and is paper, and sent to a centralized laboratory, where
not accompanied by an elevation in serum antimi- blood is eluted from the paper and analyzed for
crosomal or antithyroglobulin antibody levels, nor concentration of TSH (in some locations T4
is there an alteration in thyroid function from nor- might be determined instead). Most screening
mal. The thyroid gland slightly increases in size programs will report results by age 7 days. The
during puberty, the so-called physiologic goiter of timing of the screening is important as a sample
adolescence, but significant increases in thyroid should be collected after 24 h, sometimes diffi-
size suggest a pathologic process, and evaluation cult with the tendency to early postnatal dis-
of goiters is indicated, rather than accepting a goi- charge. A screening test is just that and is not
ter as a normal variation. reliable for diagnosis and therapy without confir-
A solitary nodule in childhood is unusual and mation. However, if the TSH value is elevated, a
worthy of careful attention and should not simply diagnosis of presumptive hypothyroidism is
be considered a routine goiter. Serious consider- made. If the result is indeterminant, the central-
ation of thyroid cancer must arise if a solitary ized laboratory in some states might offer a
nodule is noted (see later). A thyroid scan may be repeated sample. However, a high value of TSH
used to determine whether the nodule is hot and on screening should suggest the wisdom of
functional or cold and nonfunctional. A cold nod- immediately obtaining a serum FT4 and TSH by
ule is particularly worrisome, as it more likely venipuncture in preparation for instituting ther-
indicates neoplasia although functional nodules apy rapidly. If the TSH is low, it will not be
can occur in neoplasia (see later). detected on TSH-screening programs, and the
diagnosis of secondary or tertiary hypothyroid-
ism will be missed; in most cases, these diagno-
Hypothyroidism ses are accompanied by other deficits of the
hypothalamus or pituitary, and the diagnosis will
Congenital Hypothyroidism (CH) be indicated by these other conditions such as
growth hormone or ACTH deficiency, which
The detection and treatment of neonates with may lead to neonatal hypoglycemia or even
hypothyroidism should be considered a pediat- hypoglycemic seizures (see later). Since it is
ric emergency (Table 6.2). If therapy is not possible that screening tests may not be per-
begun soon after birth, developmental delay formed in a newborn or may not pick up all cases
will result. The goal of treatment is to com- of congenital hypothyroidism, the clinician
mence immediately upon diagnosis. Neonatal treating newborns must retain a high index of
hypothyroidism is a relatively common disor- suspicion for any other signs of hyperthyroidism
der, with an incidence close to one in 3,000 live that may develop.
births. CH is more common in Asian, Native It is important to view the normal develop-
American, and Hispanic populations, while mental physiology of the fetus and newborn to
Caucasians and African Americans have a lower understand the pitfalls of the neonatal screening
prevalence. It is more common in older mothers program and interpretation of thyroid function in
and in premature infants. the days and weeks after birth (read above). To
Because of the difficulty in making a clinical repeat, after delivery, serum TSH quickly
diagnosis early enough to salvage mental func- increases and peaks at 30 min; T4 is secreted in
tion based on the history and physical examina- response and increases by 24–72 h to values
tion alone, routine neonatal screening has been equivalent to those found in adult hyperthyroid-
instituted in the USA and in most countries. In ism, with a slow decrease toward adult normal
fact congenital hypothyroidism is the most com- values over the following weeks, months, and
mon disease for which neonatal screen is per- years. Serum T3 exhibits a primary increase in
formed. In most cases, samples of blood are the hours after birth and exhibits a secondary
collected from a heel stick, placed on a filter increase at 24–72 h due to the increase in T4; RT3
Table 6.2 Etiologies of congenital hypothyroidism
Disorder Subtypes Prevalence Goiter Free T4 TSH Other features Inheritance
Thyroid dysgenesis 1:3,000–5,000 No Low High May be agenesis, hypoplastic, or ectopia Mostly sporadic, but
3 % have gene mutations
Thyroid dyshormonogenesis 1:40,000
Iodine-transport defect Yes Low High Mutation of gene for sodium/iodide AR
symporter. Treated with extra iodide
Organification defect 1:40,000 Yes Low High Mutation of thyroid peroxidase. Positive AR
perchlorate discharge test is diagnostic
Pendred syndrome 1:50,000 Yes Low High Defect in pendrin gene. Deaf mute and AR
positive perchlorate discharge test
Iodotyrosine deiodinase defect Yes Low High High RAIU but early discharge. High serum AR
MIT and DIT
Hypothalamic–pituitary 1:100,00 No Low Low Mostly sporadic but may
abnormality be part of AR multiple
pituitary hormone
deficiency
Familial TSH deficiency No Low Low Abnormal TSH-β gene AR
Pit-1 or Prop-1 gene No Low Low Absence of GH and Prl secretion AR
deficiency
TSH unresponsiveness Rare No Low High Mutation of TSH-receptor gene AR
Transient hypothyroidism 1:40,000
Maternal antibodies blocking No Low High
TSH receptor
Goitrogens (medication such Yes Low High
as maternal PTU or food such
as certain cabbages)
Excess or inadequate iodine, Yes Low High
e.g., endemic cretinism
Thyroid hormone resistance 1:100,000 Yes High High Mutation of thyroid-receptor gene. May be AR, AD. Sporadic
general (TSH may be normal), peripheral,
or pituitary resistance
Thyroglobulin defects 1:40,000 Yes Low High Mutation of TG gene AR
TSH thyroid-stimulating hormone, AR autosomal recessive, AD autosomal dominant, PTU propylthiouracil, RAIU radioactive iodine uptake, MIT monoiodotyrosine, DIT diiodotyro-
sine, GH growth hormone, Prl prolactin (Modified from Fisher DA. The thyroid. In: Rudolph CD, Rudolph AM, eds. Rudolph’s pediatrics. New York: McGraw-Hill; 2002:
2059–2079)
Hypothyroidism 103

decreases after birth; TBG remains relatively perature instability, lag in the time of the initial
constant after birth. episode of stooling to more than 20 h after birth,
It is essential to understand this developmental edema, and hypoactivity and poor feeding can be
physiology to properly understand the diagnosis seen in hypothyroid patients in the neonatal
of congenital hypothyroidism. A normal child at period, most or all of these findings could be
term will have serum T4 concentrations in the signs of other diseases as well and may not sug-
adult hyperthyroid range during the 24–72 h after gest the definitive diagnosis to the physician
birth, whereas a child with bona fide hypothyroid- without the aid of neonatal screening.
ism will have serum T4 either below the normal Thus neonatal screening programs were estab-
adult range or in the normal adult range. Healthy lished to diagnose patients uniformly before
premature babies, their development interrupted definitive symptoms occur. However, the signs
before the normal stage of thyroid physiologic and symptoms of congenital hypothyroidism
development at term, have a lower concentration must be kept in mind because the statewide
of T4, and premature babies with respiratory screening programs are not infallible, and a
distress syndrome or others who are small for patient may miss screening because of home
gestational age will have T4 values lower than do birth, early discharge from the hospital, long stay
normal age-matched premature children. in the neonatal intensive care unit, transfer
A late rise in TSH is found in these neonatal con- between hospitals, or even lost samples. If a sus-
ditions which will complicate neonatal screening picion exists that a newborn might have hypothy-
tests. Some programs check a second sample in a roidism, rather than waiting for the neonatal
few weeks after birth and find an increased inci- screening result, standard serum FT4 and TSH
dence of hypothyroidism due to delayed rise in determinations can be requested, and the results
TSH; this occurs most often in premature infants. may be available even sooner than those from the
Thus, the gestational age of the neonate, the screening program.
clinical condition, and the postnatal age are
important in the interpretation of neonatal Anatomical
thyroid-screening tests. Age-adjusted normal The development of the thyroid gland is described
ranges for these tests are essential information above. The etiology of congenital hypothyroid-
from any laboratory testing at these ages and ism is usually anatomical (85 % of cases): thus
conditions. athyrosis, hypoplasia of the thyroid gland (half to
The signs and symptoms of congenital hypo- one-third of cases), or a lack of descent of the
thyroidism in its classic form include large thyroid gland from its initial site of formation to
tongue, coarse facies, umbilical hernia, a combi- its normal mature location (thyroid ectopy or, by
nation of lethargy and irritability, poor growth analogy to the term used for lack of descent of the
and weight gain, short extremities with a delayed testes, “cryptothyroidism”) occur. These condi-
or high upper-to-lower-segment ratio, persis- tions were generally considered to occur in a spo-
tently open posterior fontanel, large anterior fon- radic pattern, but there are genetic disorders in
tanel, and coarse voice. Further, pericardial 1–3 % of cases (#218700 THYROID
edema can be noted on ultrasound study in infants DYSGENESIS at 2q12-q14), although there are
left untreated for a prolonged time. However, usually other findings in these cases with gene
these signs and symptoms take weeks to months mutations. The presence of thyroglobulin in the
to develop, and if suspicion of congenital hypo- serum indicates the presence of at least some thy-
thyroidism were to be triggered by physical stig- roid tissue. A 123I thyroid scan is able to demon-
mata alone, the majority of patients would strate ectopy, but an initial finding of athyrosis or
already have irreversible mental changes before hypoplasia on scan may occur because of mater-
diagnosis was accomplished and therapy began. nal blocking antibodies that crossed the placenta
Although features such as prolonged gestation and may falsely suggest a permanent anatomic
with large birth weight, persistent jaundice, tem- abnormality, when a transient condition is
104 6 Disorders of the Thyroid Gland

actually present (see earlier). Ultrasound exami- which includes neurosensory deafness (#274600
nation of the area will help determine the pres- ICD+PENDRED SYNDROME; PDS). With a
ence of thyroid tissue in those infants with TSH deiodinase defect (#274800 ICD+THYROID
receptor-blocking antibodies (TBA or thyroid- DYSHORMONOGENESIS 4; TDH4), the indi-
binding inhibitory immunoglobulin (TBII) which vidual cannot remove iodine from the DIT or MIT
are passed from the mother who has autoimmune and therefore becomes iodine deficient from
thyroid disease herself; she may have atrophic iodine loss in the urine. Other conditions include
thyroiditis which is characterized by thyroid thyroglobulin defects causing lack of coupling of
autoantibodies, often TSH blocking antibodies, MIT and DIT (#274900 ICD+THYROID
causing functional hypothyroidism, and absence DYSHORMONOGENESIS 5; TDH5) and a
of goiter). Ultrasound may indicate the presence defect in TSH receptors (#275200-
of thyroid tissue when an infant has a genetic HYPOTHYROIDISM, CONGENITAL,
iodine-trapping defect that will interfere with the NONGOITROUS, 1; CHNG). A defect in the
demonstration of the tissue on an 123I scan. transport of iodine into the thyroid gland due to
Ultrasound is not reliable in seeking ectopic mutation in the sodium/iodide symporter which
glands however. develops the large gradient between lower extra-
Endemic cretinism is found in areas of iodine cellular iodine concentrations and higher intracel-
deficiency, but with iodine supplementation of lular concentration of iodine is not always goitrous
salt, bread, and other foods, the prevalence has (#274400THYROID DYSHORMONOGENESIS
decreased worldwide, and the condition is virtu- 1; TDH1), while defects in hydrogen peroxide
ally nonexistent in the USA except in households generation (#607200THYROID DYSHOR-
where alternative food sources are used and the MONOGENESIS 6; TDH6) are not goitrous.
family avoids supplementation. Nonetheless, Three forms of resistance to thyroid hormone
iodine deficiency is still the leading cause of are reported all with a defect in the same TSH
congenital hypothyroidism worldwide, and it receptor beta gene. Generalized (central and
is all the more tragic because it is easily peripheral resistance) to thyroid hormone
preventable. (#188570 THYROID HORMONE
Severe hypothyroidism at any age can lead to RESISTANCE at 3p24.3) is associated with
myxedema, a term derived from mucinous edema delayed speech development, attention-deficit/
as described in the nineteenth century. Myxedema hyperactivity disorder associated with goiter, but
is due to protein-bound mucopolysaccharides, hyal- euthyroid status due to increased secretion of
uronic acid and chondroitin sulfuric acid, in areas of thyroid hormone. The defect is in the TR beta
the skin which leads to a swollen appearance. receptor, and the inheritance is usually autoso-
mal dominant. Other kindreds have thyroid hor-
Biochemical mone unresponsiveness or Refetoff syndrome in
Biochemical abnormalities of the thyroid gland an autosomal recessive pattern (#274300
are usually inherited in an autosomal recessive THYROID HORMONE UNRESPONSIVE-
manner and often involve the development of a NESS at 3p24.3) in which thyroid hormone and
goiter in about 10–15 % of cases of congenital TSH concentrations are high; the patients are
hypothyroidism. The prevalence of biochemical clinically euthyroid but may have exophthalmos,
congenital hypothyroidism is about 1 in 40,000. deaf–mutism, skeletal abnormalities, and goiter;
Possible diagnoses in a with goiter include there is also a mutation in the gene for the
dyshormonogenesis, such as peroxidase defects or β-portion of the thyroid hormone receptor in this
organification defects (#274500 ICD+THYROID syndrome as well as the previous one. Autosomal
DYSHORMONOGENESIS 2A; TDH2A due to dominant selective pituitary resistance to thyroid
mutation in the thyroid peroxidase gene at 2p25) hormone (#145650 THYROID HORMONE
which may be a part of the Pendred syndrome RESISTANCE, SELECTIVE PITUITARY;
Hypothyroidism 105

PRTH) leads to hyperthyroidism since the serum T3; the free forms of both hormones are
elevated thyroid hormone levels do not suppress normal, and the patient is asymptomatic.
TSH secretion from the pituitary gland. Children with nephrosis will have massive loss
of thyroid hormone in the urine and thyroid-binding
Hypothalamic–Pituitary Abnormalities globulin due to proteinuria. Some studies reported
Rarely (fewer than 1 in 100,000 live births), isolated elevated TSH values which reverted to normal with
TSH deficiency or secondary hypothyroidism is resolution of the nephrosis.
found, but multiple pituitary deficiencies accompa-
nying TSH deficiency are found more frequently in Consumptive Hypothyroidism
an incidence of 1 in 20,000–30,000. The manifesta- Neonates may have hemangiomas with excess
tions of secondary or tertiary hypothyroidism are type 3 deiodinase activity which decreases the
usually milder than primary hypothyroidism. There biological action of thyroxine by conversion to
are extremely rare familial forms of isolated TSH inactive metabolites including reverse T3 leading
deficiency (#275100 ICD+HYPOTHYROIDISM, to often severe hypothyroidism and elevated
CONGENITAL, NONGOITROUS, 4; CHNG4) TSH. There may be a positive neonatal screen
due to mutations in TSH beta-subunit or isolated for hypothyroidism. Replacement doses of thy-
TRF deficiency (tertiary hypothyroidism) roxine are many times higher than expected for
(%275120THYROTROPIN-RELEASING age due to the fact that the enzyme inactivates
HORMONE DEFICIENCY) in the TRF receptor the administered thyroxine as well as the endog-
gene. However, TSH or TRF deficiencies are seen enous thyroxine. Most patients have been
with other pituitary hormone deficiencies which reported with infantile hepatic hemangiomato-
will likely manifest the signs that will lead to the sis, but other tumors may exert the same effects
diagnosis such as hypoglycemia, microphallus, or in older individuals.
jaundice. Genetic defects leading to multiple pitu-
itary deficiencies along with TSH deficiency are Exogenous Agents
found in mutations of Pit-1, Prop-1, HESX1, LHX4, External agents administered to the mother can
or other autosomal genes leading to many associ- cause congenital hypothyroidism. Radioactive
ated defects (see Chap. 3). iodine mistakenly given to a pregnant woman
with Graves’ disease will immediately cross the
Thyroid-Binding Globulin Mutations placenta and damage the developing gland; if the
In several situations, thyroid-binding proteins are dose is less than therapeutic, total destruction of
altered and thereby alter the serum T4 and T3 con- the gland may not occur, but neoplasia may fol-
centrations. Decreased TBG, or TBG deficiency low because of the damage to the fetal thyroid
(*314200 THYROXINE-BINDING GLOBULIN gland. Methimazole or, now rarely, PTU given to
OF SERUM; TBG at Xq22.2), occurs in 1 of a pregnant woman with hyperthyroidism also
10,000 live births in a sex-linked pattern. The will cross the placenta and can cause profound
serum T4 and T3 are decreased, and the serum FT4 hypothyroidism if the maternal dose is too great;
is normal, but because serum TSH is normal, the when the PTU is cleared after delivery, the baby
child will be not detected by the newborn TSH- will recover from the hypothyroidism caused by
screening program. Most children are normal, but this suppression, but the thyroid-stimulating
mental retardation also is reported in the litera- immunoglobulin that was passed from the mother
ture. Familial dysalbuminemic hyperthyroxinemia via the placenta will begin to exert its effect,
(FDH) (*145680 HYPERTHYROXINEMIA, causing transient but potentially clinically rele-
FAMILIAL) leads to increased T4 but a normal vant hyperthyroidism.
106 6 Disorders of the Thyroid Gland

Paradoxically, excessive iodine given to the Management of a Positive Neonatal


mother can suppress thyroid gland formation, Screen
as can a deficiency of iodine. Iodine deficiency With the receipt of a screening test result suggest-
is highly prevalent in the world and leads to ing congenital hypothyroidism, another set of
goiter, cretinism (this word is not to be used in serum samples for confirmatory T4 or free T4 and
discussion with families of course), poor TSH should be immediately drawn, as the results
growth, and hypothyroidism. Cretinism may of a screening test require proof. If the initial
present with neurological signs including TSH was quite high, treatment with thyroxine
developmental delay, mutism, and spasticity or, may be started while awaiting the results of the
with the myxedematous form, with myxedema confirmatory tests.
and more moderate developmental delay and A nuclear medicine thyroid scan may be help-
growth failure. ful if the expertise is available but is subject to
limitations noted above. A large gland on scan
Transient Congenital Hypothyroidism suggests dyshormonogenesis, a small or ectopic
and Variants gland an anatomic defect, but an absent gland can
Transient congenital hypothyroidism is seen be due to disease such as TSH or TRF deficiency
when mothers are exposed to excess iodine and or iodine symporter mutation but also may be
antithyroid medications for hyperthyroidism or temporary and due to maternal blocking antibod-
transfer antithyroid antibodies to their fetus. Rare ies. Maternal blocking antibodies are determined
cases of heterozygous mutations in DUOXA2 by a thyrotropin binding inhibitory immunoglob-
lead to transient CH. ulin or TBII assay. Urinary iodine determination
Transient hyperthyrotropinemia, in which a if low indicates iodine deficiency and if high
child will have normal serum T4 concentrations for indicates excess iodine exposure in the mother;
age but a slightly elevated TSH (often in the range both can cause a positive neonatal screen.
of 20–30 mIU/mL), may be confused with primary Measurable serum thyroglobulin will indicate
hypothyroidism. The natural history of such the presence of thyroid tissue, but absence will
patients is a decrease in the TSH concentrations to indicate the lack of thyroid tissue.
normal by age 1–2 years in the absence of treat- A bone-age determination of the knee and foot
ment. Children with laboratory values characteris- is useful in some cases if the infants is shielded
tic of transient hyperthyrotropinemia should be with lead; the distal femoral epiphysis calcifies at
watched for evidence of decreasing thyroid func- 36 weeks of gestation, the proximal tibial epiphy-
tion in case they have bone fide hypothyroidism, sis at 38 weeks, and the cuboid epiphysis of the
instead of benign transient hyperthyrotropinemia. feet at term. Although most newborns don’t
Patients with neonatal hypothyroidism will in some require this confirmation, a bone age of the leg and
cases maintain a normal serum T4 concentration at foot can be determined at the time of receipt of the
the time of the newborn screening at the expense of screening results, and the finding of delayed lower
an elevated TSH via compensated hypothyroidism; extremity bone age for gestational age may
most will be detected, but some will have normal strengthen the impression of congenital hypothy-
serum TSH at birth and for some time thereafter roidism (a hand bone age is not useful at this age).
until the inadequate thyroid gland fails. An ectopic Patients with congenital hypothyroidism may have
or hypoplastic thyroid gland that, although func- epiphyseal dysgenesis or irregular multifocal cal-
tional enough at birth to allow a normal newborn cification on radiograph when the epiphyses do
screen, decompensates with time, and true hypo- appear after the institution of treatment.
thyroidism develops; these cases may present with Treatment of congenital hypothyroidism
apparent transient hyperthyrotropinemia. Thus should begin immediately after the diagnosis is
elevated serum TSH is a worrisome finding; the established, as long as confirmatory tests beyond
differential diagnosis in this case should be made the neonatal screening tests are obtained first. In
by an experienced pediatric endocrinologist. those cases in which the screening results show
Hypothyroidism 107

Table 6.3 General starting doses of thyroxine by age in up of growth rate, serum TSH and thyroxine, or
micrograms/kg: all must be adjusted by clinical state and
free thyroxine at frequent intervals is essential to
serum thyroxine (and TSH after the newborn period)
make sure therapy is continuing and effective
Age Doses of thyroxine (μg/kg)
especially in the first 2–3 years. The American
0–3 months 10–15
Academy of Pediatrics recommends testing chil-
3–6 months 8–10
dren every 2–3 months for the first 6 months, 2–3
6–12 months 6–8
months until 3 years old, and 6–12 months there-
1–5 years 5–6
after. Lab values should have free T4 or total T4 in
6–12 years 4–5
the upper ½ of the range and TSH between 0.3
>12 years 2–3 (and lower with
completion of pubertal and 5 (or some say 0.3–2) mIU/mL. There is con-
development) tinued evaluation of the best dose of thyroxine to
TSH thyroid-stimulating hormone be used and how to monitor the dosage. There are
From various sources including prescribing information conflicting reports as to whether slight overtreat-
for levothyroxine (synthroid) from Abbott Laboratories ment is harmful. Most agree that rapid normal-
ization of free thyroxine values is appropriate.
an extremely high TSH, treatment can be started If a patient with diagnosed primary hypothy-
even before the confirmatory results return, as it roidism has not had an elevation of the serum
will be most unlikely that a mistake in diagnosis TSH to abnormally high values during treatment,
has occurred. Presently, recommended dosage of which would have demonstrated a permanent
synthetic thyroxine is 10–15 μg/kg for the new- impairment of thyroid function, and has no
born (a dose of 37.5–50 μg is the usual daily dose known anatomic defect in the thyroid gland, a
in term newborns) and less thereafter, 3–5 μg/kg careful challenge off medication may be tried, at
until a total dose of 100–150 μg is reached at the age 3 years of age to see if they have a transient
maximum in a teenager (Table 6.3). Thyroxine is defect. Such a challenge can only be carried out
most widely available in tablets and is crushed in those patients that have reliable families and
and administered in a small amount of formula, can be counted on to return for follow-up; if these
water, or breast milk in the newborn. While fast- criteria are met, thyroxine can be discontinued
ing is the best time to administer thyroid medi- for 2–4 weeks, with serum T4 and TSH measured
cines, this is not possible in the newborn; at least at 1, 2, 4, and 6 weeks to determine thyroid func-
avoiding extra calcium, iron preparations, or soy tion. Most thyroid-dependent brain growth is
will increase the likelihood that the medication completed by 3 years, so that the test is safer at
will be absorbed adequately. The chronologic this age than at an earlier age, but it is not advis-
relationship of medication to diet should remain able to leave a hypothyroid child off thyroxine
consistent for compatible longitudinal lab results. for long intervals, even at this age. If elevated
Liquid thyroxine is available for parenteral use TSH is seen confirming permanent hypothyroid-
but is quite expensive; it has been used rarely for ism, the trial off therapy is over and the child
patients who are unable to take medications deserves renewed and continued thyroxine ther-
orally. Compounding pharmacies cannot make a apy; if no elevation of TSH occurs by 6 weeks off
safe suspension that allows the same dosage per thryoxine, the child is normal.
administration, and this method is to be avoided. Neonates with congenital hypothyroidism may
T3 is not used for the treatment of hypothyroid- have elevated TSH concentrations for months or
ism, nor is a combination of T4/T3 as lay websites even years after the onset of appropriate therapy
may suggest. There is no indication for desic- because of a persistent abnormality of feedback
cated thyroid pills although naturopaths may sug- suppression; attempting to suppress these TSH
gest it. Children must be followed up carefully, concentrations to normal levels may lead to exces-
with determination of thyroxine or free thyroxine sive thyroxine dosage. Thus the goal of therapy
at 2 weekly intervals as treatment starts until should be to maintain a normal serum thyroxine
therapeutic range is reached. Continued follow- or free thyroxine concentration at the upper half
108 6 Disorders of the Thyroid Gland

of the normal range for age rather than to suppress delay. Prenatal therapy of fetuses in families
the TSH to normal values for age at this early age. with proven genetic hypothyroidism is reported.
Significant overtreatment with thyroxine will This is presently considered an experimental pro-
cause advancement of the bone age and can lead cedure. While there is evidence (still controver-
to craniosynostosis and can decrease intellectual sial) that untreated or poorly treated maternal
attainment. Further, the administration of full hypothyroidism is correlated with lower intelli-
treatment dosages of thyroxine to infants with gence in offspring who do not have hypothyroid-
congenital hypothyroidism that were untreated for ism, there is no confirmatory evidence of the
a period of months may cause the onset of conges- benefit of treating the mother with thyroxine
tive heart failure because of the rapid mobilization during pregnancy. This is in distinction to the
of fluid accumulated during the myxedematous definite improvement in fetal outcome if iodine
state; in such children with late onset of therapy, it deficiency is treated during pregnancy.
is preferable to work up to the full dose over a
period of 7–14 days. Of course, excessively low
dose of thyroxine for a long time may lead to a Acquired Hypothyroidism
poorer CNS result, so this titration of the dose
upward cannot be a prolonged process. Acquired hypothyroidism is not a rare disease.
Sick premature infants may have low TSH It is estimated that this group of disorders occurs
and thyroxine values due to their immature devel- in over 1 % of school-age children.
opment. Neonatologists have considered thyroid Childhood hypothyroidism starting after age
supplementation for severely premature infants 2–3 years does not carry the risk of permanent
with no evidence of thyroid disease other than mental retardation, as does untreated neonatal
these lab values. Clinical studies have not shown hypothyroidism, but temporary, reversible behav-
consistent benefit, and this is not recommended ior changes are frequent. Mild congenital abnor-
practice. Likewise there is no evidence of benefit malities, such as an ectopic thyroid gland, may
in treating infants with the sick euthyroid syn- first manifest in the childhood period, although
drome of low T4, T3, and TSH with thyroxine. most will have been diagnosed by moderate ele-
Patients treated early and appropriately with vations of the TSH level in the newborn screen-
thyroxine are likely to have normal intelligence ing programs. The addition of iodine to food in
according to ever accumulating data. Parents should the USA has effectively eliminated endemic
be reassured about the expected normal outcome as iodine-deficient goiter, although children born in
the institution of neonatal thyroid screening and many other countries are still at risk as are those
neonatal treatment has markedly decreased the whose parents insist on “natural” salt or other
morbidity of this common condition. In only very foods without iodine added.
rare cases, even if therapy began in the first week of Symptoms of classic hypothyroidism include
life, severe intellectual impairment is reported. cold intolerance, constipation, and some weight
The brain is dependent upon the supply of thy- gain due to myxedema. In spite of common per-
roxine which is deiodinated locally to produce ceptions that hypothyroidism is responsible for
T3, so if a fetus is normal even if the mother is significant obesity, the weight gain does not
thyroid deficient, the T3 supply to the brain increase to the extent of severe obesity. Skin is
should be acceptable in most cases. Likewise if a dry, and the hair is brittle and even sparse in
fetus is hypothyroid and the mother is normal, a hypothyroidism. Long-standing primary hypo-
small amount of thyroxine that passes the pla- thyroidism may lead to enlargement of the pitu-
centa from mother may provide adequate sub- itary gland due to increased TSH secretion:
strate to allow appropriate levels of T3 in the fetal erosion of the sella turcica may appear to indicate
brain. However, lack of maternal thyroxine in the the presence of a tumor on radiograph, so thyroid
face of fetal hypothyroidism leads to a profound function should always be evaluated in the face
deficiency of T3 leading to severe developmental of such a finding, if otherwise unexplained.
Hypothyroidism 109

Hypothyroidism causes poor postnatal growth conjunctivitis, dental enamel hypoplasia, chronic
and results in short stature. Long-bone growth is active hepatitis, cholelithiasis, asplenia, malab-
diminished, and the upper-to-lower-segment sorption, diarrhea, chronic atrophic gastritis,
ratio (see Chap. 5) is decreased for age, leading vitiligo, ectodermal dystrophy, alopecia, insulin-
to immature proportions. The bone age is delayed, dependent diabetes mellitus, transient, isolated
often significantly. Puberty and menarche is hypoaldosteronism, and pernicious anemia.
delayed or absent in teenage patients. Treatment Type 2 (269200 SCHMIDT SYNDROME)
will increase growth rate to values often higher consists of hypo- or hyperthyroidism, Addison’s
than the average for age leading to “catchup disease, type 1 diabetes mellitus, but not hypo-
growth.” In addition, treatment of acquired hypo- parathyroidism nor candidiasis.
thyroidism will advance bone age and, if the Type 3 autoimmune polyendocrinopathy may
child is old enough, precipitate appropriate be invoked to describe Hashimoto thyroiditis
pubertal development. However, if the condition with one of the other autoimmune endocrine
is untreated for a long period, adult height will be organ failures with the exception of Addison’s
compromised even with appropriate treatment. disease which is not included in type 3. Thus
other features of the type 3 syndrome may include
Autoimmune Thyroid Disease: type 1 diabetes mellitus, celiac disease, hypogo-
Hashimotos Thyroiditis nadism, sarcoidosis, Sjogren syndrome rheuma-
Hashimoto thyroiditis or chronic lymphocytic toid arthritis, vitiligo, and alopecia.
thyroiditis or autoimmune thyroid disease Clinical characteristics of Hashimoto thyroid-
(*140300 HASHIMOTO STRUMA at 8q23-q24) itis include goiter, minimally to severely elevated
is responsible for the majority of cases of acquired TSH concentrations, and frequently a family his-
hypothyroidism. This autoimmune disorder is tory of thyroid disease. A characteristic pebbly
often found in the same family group as Graves’ surface to the gland is felt because of accentua-
disease. In Hashimoto thyroiditis, there is forma- tion of the normal follicular structure of the
tion of unsuppressed clones of thymus-dependent gland. A positive titer of antithyroid peroxidase
lymphocytes directed against the thyroid gland, antibodies or antithyroglobulin antibodies or
causing lymphocytic infiltration of the gland. B both are helpful in diagnosis; children more fre-
lymphocytes, either through interaction with T quently have the former antibodies, whereas
lymphocytes or because of stimulation linked to adults more usually have the latter. Lymphocytic
the damaged thyroid cells, produce antithyroid infiltration of the thyroid gland occurs, with the
antibodies, antithyroglobulin, or antithyroid per- formation of germinal centers on histologic study
oxidase antibodies (previously called antimicro- (there is no need to obtain tissue for histology for
somal antibodies) that are characteristic of the diagnosis however). There is no reason to per-
disorder and are a cornerstone of the diagnosis. form a thyroid scan or uptake in a well-
Hashimoto thyroiditis is frequently associated documented case of Hashimoto thyroiditis.
with other disorders. Autoimmune polyglandular Elevated serum TSH and decreased FT4 indi-
diseases may include Hashimoto thyroiditis: cate obvious primary hypothyroidism. Lower but
Type 1 (#240300 ICD+AUTOIMMUNE slightly elevated TSH concentrations with nor-
POLYENDOCRINE SYNDROME, TYPE I, mal serum FT4 present a problem. As noted
WITH OR WITHOUT REVERSIBLE above, TSH values in childhood may be consid-
METAPHYSEAL DYSPLASIA; APS1) may be ered normal by some evidence. Normal children
inherited in an autosomal dominant or autosomal who have TSH values up to 7.5 mIU/L in a study
recessive manner and consists of two of three with years of follow-up demonstrates no further
major conditions hypoparathyroidism, Addison’s elevation in most cases and often decreases to
disease, and mucocutaneous candidiasis, which lower levels in children. However, if the TSH is
might be the first sign noted. Other findings may definitively elevated, the treatment is clear: the
include hypothyroidism, hypogonadism, kerato- administration of thyroxine until the TSH is sup-
110 6 Disorders of the Thyroid Gland

pressed to normal and the serum FT4 increases to removed. Internationally, iodine deficiency is the
or stays at normal. If the TSH is not elevated and most common cause of acquired hypothyroidism
the goiter is minimal, the patient may be regu- as endemic cretinism (above). Excess iodine
larly evaluated for further deterioration in thyroid intake (e.g., from kelp given by a naturopath,
function during the following months and years. medications containing iodine, radiographic
If the goiter is noticeable, thyroxine may be given dyes, and/or topical from antisepsis therapy such
for a period of 3 months to see if the goiter can be as Betadine for an open umbilical wound) may
diminished; if the small goiter does not change, suppress thyroid function and temporarily lead to
thyroxine may be stopped and the patient fol- hypothyroidism, but will later escape due to the
lowed up until the thyroxine treatment may Wolff–Chaikoff effect. Certain drugs impair thy-
become definitively necessary. roid function such as lithium which can cause
Rarely children are born with only one lobe of hypothyroidism and goiter or amiodarone which
thyroid gland, usually the left. This tissue may may cause either hypothyroidism or hyperthy-
undergo compensatory hypertrophy, and it has roidism. Infiltrative disease from Langerhans cell
the usual chance of developing Hashimoto thy- histiocytosis or lymphoma or hemochromatosis
roiditis, causing a presentation of hypothyroid- due to iron infiltration of the thyroid gland due to
ism with one-sided goiter. transfusions for thalassemia or cystinosis may
Annual screening is recommended for Down cause primary hypothyroidism. Infiltrative dis-
syndrome, Turner syndrome, and Klinefelter syn- eases such as lymphoma and Hodgkin’s disease
drome, all of whom are predisposed to the devel- and their treatment can affect thyroid function as
opment of autoimmune hypothyroidism. rarely can a teratoma. Treatment for hyperthy-
Remarkably, premature sexual development roidism (e.g., surgery or radiation) even if ini-
occurs in some children with untreated primary tially leading to euthyroid status will often later
hypothyroidism which is identified as the Van lead to hypothyroidism. A liver hemangioma caus-
Wyk–Grumbach syndrome; girls have breast ing elevated activity of type 3 deiodinase that pref-
development, and boys have enlargement of tes- erentially converts T4 to reverse T3 and T3 to T2,
tes and possible slight enlargement of the penis, thereby decreasing available thyroid function, will
but usually don’t demonstrate premature devel- lead to acquired hypothyroidism; sometimes the
opment of pubic hair. Galactorrhea may be found hypothyroidism is diagnosed first and leads to the
because of increased secretion of prolactin in pri- diagnosis of the hemangioma. Some hemangioma
mary hypothyroidism, as TRF stimulates prolac- are reported to shrink with glucocorticoid therapy
tin as well as TSH release (see Chap. 9a). resolving the inactivation of thyroid hormone.
With improvement in oncology care, more
Other Causes for Acquired and more children survive cancer. If there was
Hypothyroidism radiation to the head and neck, hypothyroidism
Other causes for acquired hypothyroidism may develop in close to one-third, and all are at
include a thyroglossal duct cyst (188455 risk for the development of thyroid carcinoma.
THYROGLOSSAL DUCT CYST, FAMILIAL) Chemotherapy may affect thyroid function as
for the rare genetic form in the midline of the may infiltrative conditions such as Hodgkin’s
neck. While usually non or poorly functional, it disease. Thus, laboratory testing with free T4 and
may contain the only functioning thyroid tissue TSH is indicated and ultrasonography to detect
available to the patient; if any midline congenital the formation of nodules.
defect of the neck is removed surgically, it may Any disease of the hypothalamic–pituitary
contain a thyroglossal duct cyst; the patient must area may decrease TSH secretion, leading to sec-
be evaluated for the development of hypothyroid- ondary or tertiary hypothyroidism (see Chap. 3).
ism, or, alternatively, the patient should have a Abnormalities of TBG may be congenital (as
thyroid scan to confirm the presence of remaining above), but also may be acquired. Thus, increased
thyroid tissue which will function after the cyst is TBG binding of thyroid hormones occurs with
Hypothyroidism 111

estrogen treatment or pregnancy or is the result of on weight, up to a maximum of 100–150 μg (see


an inherited condition; decreased TBG binding Table 6.3); older children require less thyroxine
of thyroid hormones occurs with androgen ther- per kilogram body weight than is used in a neo-
apy, protein-losing conditions, and an X-linked nate with hypothyroidism. Unlike some neonates
genetic condition (see earlier). If free T4 is nor- with congenital hypothyroidism, older children
mal, thyroxine therapy may not be needed. There can safely have the dose titrated until the serum
is significant and sometimes contradictory litera- TSH is suppressed to normal. In the most severe
ture of the effects of antiepileptic drugs including cases with severe myxedema or even pericardial
phenytoin (Dilantin), phenobarbital, valproate, effusion, the dose must be slowly increased.
carbamazepine, oxcarbazepine, and levetirace- Pseudotumor cerebri is rarely encountered after
tam; there are reported changes in thyroid func- the initiation of treatment in severe cases and
tion in some but no definite evidence that sometimes even before treatment commences. No
thyroxine therapy is necessary just due to the proven advantage is found in T3 or T4/T3 nor des-
medication use. Clinical correlation is necessary. iccated thyroid treatment over thyroxine treat-
ment. Treatment with thyroxine should return the
Euthyroid Sick Syndrome child to the baseline state. Thus if the child was
Euthyroid sick syndrome presents with abnor- hyperactive before hypothyroidism developed,
mal thyroid laboratory findings in an individ- treatment may return the child to hyperactivity
ual with nonthyroidal illness without any and school work may suffer. Parents must know
disorder of the hypothalamic pituitary thyroid this in advance and understand the treatment is
axis. After the illness resolves the laboratory necessary and support in school must follow.
tests revert to normal. Characteristically Parents must understand that once the diagnosis
decreased serum T3 and serum T4 with possibly of acquired hypothyroidism is established, the
low TSH is found with elevated reverse T3 as is treatment will likely be lifelong.
found in the fetus; some call this the low T3 The half-life of thyroxine is 5–7 days.
syndrome. Thyroid treatment is usually not Stabilization after initiation of thyroxine treat-
necessary in this condition since the condition ment or a dose change usually takes 5–6 weeks to
resolves with treatment of the underlying occur, so in most cases laboratory tests should be
disorder. timed to this interval. T3 is not usually used for
therapy; the half-life of T3 is only 1 day. This can
Diagnosis of Hypothyroidism be used to advantage when a nuclear medicine
Diagnostic techniques are outlined above in indi- scan has to be performed. The patient can be
vidual disorders. It is important for the provider switched to T3 for 4–6 weeks and the T3 discon-
to realize that there is a normal variation in thy- tinued the day before the scan is performed; this
roid function tests which extend beyond the labo- avoids unnecessary malaise of the hypothyroid
ratory reference intervals in some cases and to be state which would last 6 weeks if T4 was discon-
aware that subtle changes from the laboratory tinued without any thyroid hormone replacement
standards may not be grounds for treatment. while waiting for the scan.
Likewise some individuals in the population have Overtreatment is detrimental as is lack of
antithyroid antibodies with no apparent disease treatment. Excessive thyroxine dosage can
and no need for treatment. Details of diagnosis advance bone age and lead to decreased intellec-
are found above for various conditions. tual development. Even normal doses of thyrox-
ine have been reported to rarely cause
Treatment of Hypothyroidism pseudotumor cerebri in children.
Acquired hypothyroidism is treated with thyrox- In the absence of diagnosed hypothyroidism,
ine in a variable dose, depending on age and based thyroxine is not to be used as a “weight loss drug.”
112 6 Disorders of the Thyroid Gland

Hyperthyroidism may be required for the 12 weeks or so until the


stimulatory antibodies decrease. In addition,
Neonatal Hyperthyroidism Lugol solution may be given to inhibit thyroxine
Maternal thyroid-stimulating immunoglobulins secretion and glucocorticoids may be given in
freely pass the placenta and can have significant high dosage to limit T4 and T3 conversion.
effects on the fetus. These antibodies may last for Once thyroid levels decrease the child is sus-
years in the mother after she has been treated for ceptible to the effects of hypothyroidism on brain
hyperthyroidism and may affect her fetus even development due to overtreatment with thyroid
though she is asymptomatic due to surgery or lowering agents. Close monitoring and augmenta-
radioactive iodine therapy administered years tion of low serum T4 with thyroxine as necessary
earlier. Only a minority of offspring of a mother during this period and the antithyroid medications
with Graves’ disease will have signs of hyperthy- should be weaned down as soon as feasible.
roidism. Fetal hyperthyroidism may be indicated Mothers with hyperthyroidism may take
by tachycardia and IUGR on ultrasound. Maternal methimazole while breast-feeding. Current guide-
hyperthyroidism requires referral to a high-risk lines suggest that the amount of methimazole in
obstetrics center with appropriate therapy for the the breast milk is too low to cause significant
mother. If the maternal dose of antithyroid medi- effects as long as the mother’s dose is no more
cation is excessive, the child may be born pro- than 20 mg per day. It is suggested that she take
foundly hypothyroid, with goiter, and can even the medication immediately after breast-feeding.
have severe respiratory insufficiency because of However if the child has any suggestion of liver
pressure on the trachea exerted by the large goi- disease or blood dyscrasia, evaluation is war-
ter. At birth an infant of maternal hyperthyroid- ranted since these can be caused by methimazole
ism may have a positive screen for congenital in rare cases.
hypothyroidism if the mother was treated with
PTU or methimazole that pass through the pla- Older Children
centa to the child. After the medication effect is Hyperthyroidism in pediatrics is usually due to
eliminated in a few days, neonatal hyperthyroid- Graves’ disease (#275000 GRAVES DISEASE),
ism may develop. If there are extreme manifesta- an autoimmune disease that may coexist with
tions in some, mortality is possible. The newborn Hashimoto thyroiditis (Table 6.4). The incidence
may develop goiter, tachycardia, elevated blood of Graves’ disease is lower than Hashimoto thy-
pressure, failure to gain weight, flushing, and roiditis in childhood, about 1:10,000 for Graves’
even exophthalmos. Congestive heart failure due disease. Girls are more often affected than boys
to supraventricular tachycardia and hepato- (5:1) with Graves’ disease. Human leukocyte
splenomegaly may occur. Methimazole therapy antigen (HLA)-DR3 and HLA-B8 predispose

Table 6.4 Etiologies of hyperthyroidism


Disorder Free T4 TSH RAI uptake Other features
Autoimmune Graves’ disease High Low Early elevation Thyroid-stimulating immunoglobulin
or TSH receptor antibodies positive
TSH-secreting pituitary tumor High High NA Look for pituitary tumor on MRI
Pituitary T3 resistance High High NA Increased TSH is secreted because of
resistance to T3 feedback
Nodular hyperthyroidism High Low Hot nodule (nodule takes RAI uptake demonstrates the activity
up 123I, but the rest of the of the nodule
thyroid gland does not)
TSH receptor-activating High Low NA Could be isolated or part of the
mutations McCune–Albright syndrome
RAI radioactive iodine, SH thyroid-stimulating hormone, MRI magnetic resonance imaging, NA not applicable
Hypothyroidism 113

Caucasians to Graves’ disease. The peak age at affected adults (this is not to be confused with the
onset is near adolescence. Thyroid-stimulating myxedema of profound hypothyroidism, which
hormone receptor antibodies (TR-Ab) and shares a term, but means protein-bound muco-
thyroid-stimulatory immunoglobulin (TSI) (vari- polysaccharides deposition in areas of the skin
ous laboratories use various abbreviations for this which leads to a swollen appearance). Signs of
same test or similar ones, so check with the labo- Graves’ disease also include systolic hyperten-
ratories for the tests offered; most commercial sion and velvety (smooth) and moist skin. Deep
tests do not differentiate between stimulating tendon reflexes are brisk, even to the point of clo-
(TRS-Ab), neutral, and blocking antibodies nus especially in the ankle jerks. Supraventricular
(TRB-Ab)) are directed toward the TSH receptor, tachycardia may be a presenting symptom and
leading to autonomous thyroid function and lead to an admission to a pediatric ICU before the
hyperthyroidism with suppression of TSH. Other diagnosis of hyperthyroidism is considered or
antibodies displace TSH from the TSH receptor established.
but do not stimulate the thyroid gland (these are Before establishing a diagnosis of Graves’
thyroid-binding inhibiting immunoglobulins disease, it is important to consider factitious
(TBIIs); these vary as TBII from patients with hyperthyroidism. Thyroid hormone is readily
Hashimoto thyroiditis decrease thyroid function, available in the medicine cabinet of many (often
while TBII in Graves’ disease sometimes block middle-aged individuals) some diagnosed with
the stimulatory antibodies and sometimes do bona fide hypothyroidism and others erroneously
not). T3 is produced more efficiently in Graves’ diagnosed as such. The wide availability of thy-
disease than in the normal state, and both T4 and roxine may lead to toxic ingestion of parent’s
T3 exceed the TBG capacity, so that FT4 and FT3 thyroxine by an infant or even Munchausen’s by
increase. In some cases, the T4 is normal, but the proxy by the administration of thyroxine to a
T3 is elevated; in the diagnosis of hyperthyroid- child.
ism, both should be measured, as both must be A hot nodule rarely may be active enough to
suppressed during therapy. Serum TSH is sup- cause hyperthyroidism in a child.
pressed in Graves’ disease because of the autono- Hamburger thyrotoxicosis is due to ingestion
mous formation of thyroid hormones. of ground beef in which the bovine thyroid asso-
Weakness, increased pulse rate, emotional ciated with neck muscles was included in the
lability or other behavior changes (sometimes to serving.
the point of apparent psychiatric disease and
remarkable personality changes to the point of Diagnosis
engaging in illegal activity out of character for Laboratory findings in hyperthyroidism are ele-
the patient), hyperactivity, and lack of attention vated serum T4 and T3 (both in either the total or
span, weight loss, and diarrhea are characteristic free determination) with nondetectable or sup-
findings. Subtle or significant goiter may be pressed TSH. If the TSH is not suppressed, the
found, possibly with a bruit over the thyroid patient may have a TSH-secreting condition as
gland. Exophthalmos may occur because of infil- the etiology for hyperthyroidism, an exception-
tration of glycosaminoglycan in the posterior ally rare occurrence in pediatrics. A positive
area of the orbit. Increased autonomic tone can TSH receptor antibody or TSI titer may be
cause lid retraction and “stare.” These two found (but check with the specific laboratory
ophthalmologic conditions may at first look simi- for the designation of this test, as noted earlier).
lar but should be noted separately. Pretibial myx- Often positive titers of antithyroglobulin and
edema, sometimes called Graves’ disease antimicrosomal antibodies are seen; if the titers
myopathy or peau d’orange (orange peel) skin, of antimicrosomal and antithyroglobulin anti-
presents as a waxy, discolored induration of the bodies are very elevated, the patient may have
skin on the anterior aspect of the lower legs in the hyperthyroid phase of Hashimoto thyroid-
some patients, although it is more common in itis in which preformed thyroid hormone is
114 6 Disorders of the Thyroid Gland

released (often called Hashitoxicosis), which Treatment of Hyperthyroidism


may be short lived and not require a permanent There is a chance of remission in children and
type of treatment; thus follow-up is very impor- adolescents with Graves’ disease. The likelihood
tant. Thyroid growth-stimulating antibodies of remission increases with milder abnormalities
and thyrotropin receptor-blocking antibodies of laboratory tests at presentation, resolution of
are found, as well as lymphocytic infiltration of the hyperthyroid state within 3 months of starting
the thyroid gland. treatment, and older age at onset. About 20 %
It is usually not necessary to perform a thyroid of affected children go into remission for every
scan or uptake if the constellation of symptoms 2 years of treatment, but the rate of remission pla-
and laboratory results are classic. If, however, a teaus in about 8–10 years with a total of about
scan is used, an early (4-h) determination of 50–75 % achieving a remission overall in various
uptake should be made before turnover of the series. Three types of treatment for hyperthyroid-
radioactive tracer occurs in this condition of ism are used: medication, radiation therapy, and
increased thyroid metabolism. The results of a surgery.
RAI scan in hyperthyroidism will be increased Knowing the factors which enhance the
uptake, while in Hashitoxicosis uptake will be likelihood remission help determine whether a
decreased. progression to permanent therapy such as surgery
A complete blood count (CBC) is obtained or radiation is best if medication is being consid-
to detect changes in the white blood cell ered for the first treatment modality.
(WBC) count due to the hyperthyroid state; the
WBC count may already be decreased in Medication
untreated hyperthyroidism or may be decreased Methimazole is the only medical therapy now rec-
later by medical therapy for hyperthyroidism. ommended for children and adolescents in the
Liver function should be evaluated before USA. Methimazole blocks organification of iodine
starting the thyroid medication: for reference and decreases T4 to T3 conversion. PTU, previ-
ALT > 25U/L (boys) and > 22U/L (girls) are ously recommended, now has a black box warning
considered normal but higher levels may be that it should not be given to children (nor adults)
found in untreated hyperthyroidism. due to risk for liver cirrhosis which may lead to the
Propylthiouracil (PTU) is no longer used on a ultimate need for a liver transplant. Methimazole
regular basis in pediatrics but in rare occasions is given in doses of 0.2–0.5 mg/kg per day (the
might have to be invoked for brief therapy during widest reported range for dosage of methimazole
preparation for surgery should methimazole is 0.1–1.0 mg/kg per day) which usually comes
cause undue side effects. Determination of anti- out to 15–45 mg/day for a teenager. It is generally
nuclear antibody (ANA) titers should be obtained divided into 2–3 doses per day, but some will
if PTU is to be used as treatment to ensure that a achieve control with a dose given once or twice per
positive antibody determination, a finding some- day. The aim is to cause a sufficient suppression of
times reported in the untreated hyperthyroidism, the autonomous thyroid function to bring about a
is not present before medication is given; PTU euthyroid state; this suppression of the thyroid
may seriously affect liver function or cause a gland is not a cure of the disease but rather a con-
lupus-like syndrome and carries a black box trol of the condition. The patient is followed up for
warning from the FDA. shrinkage of the goiter, an essential finding if
Some children will have elevated thyroid lev- remission is to be expected. Guidelines suggest the
els without symptoms of hyperthyroidism, which use of medication for only 1 year, but in practice
is described as subclinical hyperthyroidism. This many continue for multiple years before consider-
may persist for years before Graves’ disease ing a permanent type of therapy.
develops, and therapy may not be necessary in Side effects occur 5–20 % of the time during
the initial stage. Some may normalize. such medical therapy. Serious side effects of
Hypothyroidism 115

methimazole are not common but can be life- therapy, and thus calcium and vitamin D moni-
threatening; granulocytopenia or agranulocytosis toring and if necessary treatment are in order.
is dose dependent and usually is found in the first
100 days of use: if fever, sore throat, or general ill Surgery
feelings arise, a CBC must be checked before Total or near total thyroidectomy is an alternative
administering more methimazole. Stevens– therapy, but subtotal thyroidectomy is associated
Johnson syndrome or other rashes are reported. with a higher recurrence rate. An experienced
Arthralgia may occur and reversible cholestasis high-volume thyroid surgeon (30 cases per year)
is a risk so warnings for development of jaundice should perform the operation so that complica-
are indicated. Mild peroxidase–antineutrophil tions will be unlikely; there is a 30 % complica-
cytoplasmic autoantibodies (ANCA)-associated tion rate in children in general, but if a high-volume
vasculitis may occur later well, but the side pediatric thyroid surgeon is used, the rate
effects noted above generally occur before 1 year decreases to 4 % in a recent study. Recurrent
treatment. laryngeal nerve paralysis is an unusual but possi-
PTU was known to cause lupus-like syndrome ble complication. Transient hypocalcemia can
and hepatitis possibly leading to cirrhosis, but the follow thyroidectomy because of postoperative
switch to the use of methimazole has lessened edema of the parathyroid glands, or the condition
such risks. could be permanent if the parathyroid glands are
These drugs should be discontinued if serious removed along with the thyroid tissue. Thyroid
side effects develop, and alternative methods of storm could develop rarely during thyroid sur-
therapy should be used which include surgery gery. Preparation for surgery involves the use of
and radioactive therapy. Lack of compliance, a medical management to control the hyperthyroid
common condition in the adolescent age group, state. Some use supersaturated iodine solution
also is an indication for surgical or radioiodine (Lugol’s, 8 drops per day) for the 10 days before
therapy. Short-term carefully monitored use of surgery; the iodine will reduce the blood supply to
PTU may be considered if severe reaction to the gland. Iodine has only a temporary effect in
methimazole occurs. suppressing thyroxine secretion from the hyper-
Propranolol (2–3 mg/kg/day or 5–10 mg every thyroid gland; escape usually will occur after 2–4
6 h as a starting dose) can control symptoms of weeks (the Wolff–Chaikoff effect), so the surgery
hyperthyroidism while awaiting another mode of should be set soon after starting iodine treatment.
therapy to exert its effects and is useful in short- Although the surgeon may be able to remove the
term situations such as in preparation for surgery optimal amount of tissue to allow euthyroid func-
or in the face of thyroid storm; side effects of pro- tion after surgery, hypothyroidism may develop in
pranolol on respiratory (bronchospasm) or circu- the years after surgery because of continued scar-
latory (hypotension) function are possible, so ring and reduced function of the remaining thy-
patients must be chosen for this medication care- roid tissue, according to some reports. Thyroid
fully in case they have asthma or another condi- storm may develop during surgical therapy, espe-
tion in which propranolol is contraindicated. cially in patients not yet under control, so pro-
Studies have shown propranolol to be helpful for pranolol therapy may be needed.
long-term management in adults, but it has not
generally been used as such in children. Nuclear Medicine Therapy
Definitive types of therapy are surgery or Radioactive iodine (131I; RAI) therapy has been
radioactive iodine treatment. With successful used in hyperthyroidism for more than 50 years
treatment of hyperthyroidism even without and has proved safe in studies of adolescents who
affecting the parathyroid glands, the child may received this therapy. No thyroid cancers have
experience “hungry bone syndrome” and a ten- been detected if the dose of radioactivity is calcu-
dency toward hypocalcemia for months after lated to destroy all thyroid tissues. However,
116 6 Disorders of the Thyroid Gland

there are reports of the development of secondary HYPERTHYROIDISM, NONAUTOIMMUNE),


cancers in the tissue surrounding the thyroid so that constitutive activation leads to uncon-
gland four to five decades after radioactive iodine trolled thyroid hormone secretion. Findings
therapy, and this factor must be considered when include low birth weight, tachycardia, advanced
considering radioactive iodine therapy. No effects bone age, delayed motor development, delayed
on offspring of adults previously treated with speech development, mental retardation, sleep
RAI are noted unless of course it is administered difficulties, hyperactivity with laboratory find-
during pregnancy. Although no documentation of ings of decreased TSH, and increased free T4.
statistically increased risk is found, some still They will have no exophthalmos nor pretibial
consider it ill advised to administer RAI to myxedema.
younger children for a potentially reversible con- The McCune–Albright syndrome may dem-
dition. The Pediatric Endocrine Society sug- onstrate autonomous thyroid function because of
gested that 131I therapy may be beneficially used mutations of the G protein (see Chap. 8).
in younger patients, but it is more commonly
used now in adolescents. As one dose of RAI Thyroid Storm
may not cure the patient especially if the dose is Thyroid storm is a rare complication in children
inappropriately low, another dose may be neces- and adolescents. Thyroid storm may present with
sary. Thyroid storm could occur during RAI treat- acute onset of hyperthermia and tachycardia in a
ment, especially in patients not yet under control, patient with underlying hyperthyroidism.
so propranolol therapy may be needed here as Precipitating factors include surgery, infection,
well. pregnancy, and diabetic ketoacidosis. It may
Female patients of reproductive age must be occur during surgical or radioiodine therapy for
warned about the risks to a fetus of Graves’ dis- hyperthyroidism. Symptoms include high fever,
ease itself and of the effect of medical therapy for sweating, tachycardia, and reduced mental state
the condition. Radiotherapy for hyperthyroidism ranging from confusion to coma. Immediate ther-
must not be administered if the patient was apy is indicated for this severe condition.
pregnant. Propranolol (1–3 mg/kg orally per day, divided
Treatment of neonatal Graves is detailed into a dose every 6 h as a starting dose) can con-
above. trol some symptoms of thyroid storm. Propranolol
Hyperthyroidism rarely occurs because of a may be given intravenously at a dose of 0.01 mg/
thyroid adenoma (hot nodule) or carcinoma, sub- kg up to a total of 5 mg at a rate of less than 1 mg/
acute thyroiditis, or suppurative thyroiditis. These minute, but an intraatrial pacing catheter is a nec-
possibilities are covered elsewhere in this chapter. essary precaution because of the risk of serious
Two rare instances occur when hyperthyroid- bradycardia. Dexamethasone in a dose of 1–2 mg
ism might be found with elevated serum TSH every 6 h can reduce serum T3. Lugol’s solution
values. Selective pituitary resistance to thyroxine of concentrated iodine can be given orally if the
(145650 HYPERTHYROIDISM, FAMILIAL, patient is conscious to decrease the release of
DUE TO INAPPROPRIATE THYROTROPIN thyroid hormone from the thyroid gland; intrave-
SECRETION, autosomal dominant) leads to lack nous NaI in a dose of 1–2 g/day divided into 1/2
of feedback inhibition to T3 feedback, so TSH dose ever 12 hours may be given if the patient is
increases, and hyperthyroidism results, as the rest adult sized and is unconscious. A cooling blanket
of the body can respond to the increased secre- can help control the hyperpyrexia. Methimazole
tion of thyroid hormone even if the pituitary will not take effect for several days, but to plan
gland cannot. Further, a rare TSH-secreting pitu- for the possibly extended course of the disorder,
itary tumor might cause hyperthyroidism with 20–30 mg can be given every 6 h (by slurry, if
elevated serum TSH and FT4. necessary) in larger children. Fluid management
Hyperthyroidism may be caused by muta- must be observed, and if tachycardia causes heart
tions of the TSH receptor (#609152 failure, digitalis may be necessary.
Hypothyroidism 117

Thyroid Hormone Overdose lower likelihood of malignancy. Ultrasonographic


evidence that the nodule is a cyst makes it less
A toddler or older child may ingest excessive likely to be malignant, but carcinomas may be
amounts of an adult’s thyroid hormone. found in the walls of thyroid cysts, and the solid
Alternatively, there is the possibility that portion of a cyst is a target for diagnostic fine-
Munchausen’s by proxy may be considered in needle aspiration. The presence of enlarged
which the dose is purposefully administered. anterior cervical lymph nodes, metastases on
Close to one-third of the reported cases of thy- chest radiograph, or hoarseness all make the
roxine overdose in the USA in 2008 were in the likelihood greater that the single thyroid nodule
pediatric age group. Thyroxine takes several days is malignant.
to exert its effect due to its 5–7-day half-life, so Carcinomas of the thyroid gland are rare in
the presentation may be delayed after the expo- childhood (these constitute less than 1 % of child-
sure. Triiodothyronine however is more rapid hood cancers and less than 10 % of all thyroid
acting, and the effects will occur similar. The pre- cancers), but certain historic features increase the
sentation of thyroid hormone excess is similar to likelihood of a thyroid mass being malignant.
that of hyperthyroidism or thyroid storm. Thus, Children with thyroid Ca have a good prognosis
the patient will manifest gastrointestinal symp- for survival in spite of a higher risk of recurrence
toms including abdominal pain, nausea and vom- than in adults even if metastases may be discov-
iting, diarrhea or anorexia, nervous system ered at presentation. A history of low level irra-
findings including anxiousness, tremor, increased diation of the thyroid gland (e.g., inadvertently
deep tendon reflexes, confusion, psychosis, or while treating acne, enlarged thymus, or ring-
even seizure. Adrenergic effects of thyroid hor- worm, as was done in the past, or due to a nuclear
mone overdose include rapid pulse, palpitations, accident, as in Chernobyl) at a young age is sig-
sweating, hypertension or hypotension, and ele- nificant, and if the irradiation was done for the
vated temperature. Because of the increased met- therapy of another cancer (e.g., at the time of
abolic rate in hyperthyroidism, adrenal bone marrow transplant), the risk is higher. The
insufficiency may develop more easily if the risk of cancer is greatest if radiation occurs dur-
patient is already developing autoimmune ing early childhood, but thyroid tumors appear
Addison’s disease. Long-term exposure to ele- even decades after irradiation. The local radiation
vated doses of thyroxine will increase symptom- used for the treatment of Graves’ disease does
atology, and in addition to advanced bone age not appear to cause thyroid carcinoma, as it elim-
and craniosynostosis in younger children, weight inates all of the thyroid follicular cells as it con-
loss may occur and the patient may progress to trols Graves’ disease. Indeed, there is no record
coma. An EKG in an individual with excessive of thyroid cancer developing in children treated
thyroxine ingestion will likely show supraven- with ablative doses of radioactive iodine for
tricular tachycardia. Graves’ disease. However, a 50-year follow-up
of pediatric patients treated with radiation doses
similar to that used for Graves’ disease shows
Neoplasms increased risk of carcinoma in the tissue sur-
rounding the thyroid that was irradiated but not in
A single firm nodule of the thyroid gland is a thyroid tissue itself as it was eliminated. Staging
more ominous finding in a child than is multiple in children according to adult standards is not as
nodular goiter. The majority of solitary lesions helpful as in adults to predict future complica-
will be benign, but carcinoma is possible. Lack tions. Likewise while adult nodules are biopsied
of concentration of 123I on thyroid scan (a cold when they reach 1 cm in diameter in most cases,
nodule) increases the likelihood of carcinoma as in children biopsy should occur when nodules are
functioning nodule (warm or hot nodule) has a 0.5 cm.
118 6 Disorders of the Thyroid Gland

Many areas and cell types of the thyroid Familial Nonmedullary Thyroid Carcinoma
gland may undergo neoplastic degeneration. A Familial occurrence of papillary or follicular Ca
survey between 1973 and 2004 of children with is recognized in families with 3 or more members
thyroid carcinoma in the USA revealed that with DTC. Screening of family members at 18
83 % had papillary carcinoma, 9.5 % had fol- years of age is recommended.
licular carcinoma, and 5 % had medullary
carcinoma. Papillary Carcinoma
Papillary carcinoma may demonstrate enlarged
Medullary Carcinoma lymph nodes as well as a thyroid nodule. This
of the Thyroid (MCT) may be associated with Gardner syndrome
Medullary carcinoma of the thyroid (MCT) arises (*175100 ADENOMATOUS POLYPOSIS OF
from parafollicular C cells and is most often THE COLON; APC at any of 5q21-q22, 1p34.3-
found in the MEN 2 syndrome (see Chap. 10) 1p32.1), which is transmitted in an autosomal
(#171400 MULTIPLE ENDOCRINE dominant pattern and includes disorders of mul-
NEOPLASIA, TYPE II; MEN 2) at 10q11.2 or tiple systems including colonic polyps and other
(#162300 MULTIPLE ENDOCRINE neoplasias; follow-up is necessary in case thyroid
NEOPLASIA, TYPE IIB; MEN 2B at 10q11.2) carcinoma develops in a patient diagnosed with
as almost all cases of MEN 2 are associated with Gardner syndrome. Autosomal dominant forms
MCT. C-cell hyperplasia may precede develop- of papillary carcinoma exist (#188550 THYROID
ment of medullary carcinoma of the thyroid. CARCINOMA, PAPILLARY associated with
Isolated MCT can occur in an autosomal domi- any of 17q23-q24, 14q, 10q21, 10q11.2, 8p22-
nant condition (#155240 MEDULLARY p21.3, 1p13, 7q32-q34 loci). Abnormalities in
THYROID CARCINOMA, FAMILIAL; MTC at the ret or trk oncogenes may be found in papil-
1q21-q22, 10q11.2). Germ cell-line mutations in lary Ca.
the ret oncogene, a proto-oncogene with a tyro-
sine kinase domain on chromosome 10q11.2, are Follicular Thyroid Carcinoma
demonstrated in patients with MCT with or with- Follicular thyroid carcinoma is usually found in a
out MEN 2. Prophylactic thyroidectomy is rec- sporadic pattern but may be an autosomal dominant
ommended by 10 years or earlier in affected condition (188470 THYROID CARCINOMA,
patients with appropriate ret mutations. FOLLICULAR; FTC at 10q23.31). This neoplasm
is found less frequently in children than the other
Epithelial-Derived Differentiated two types listed.
Thyroid Cancer (DTC)
Epithelial-derived differentiated thyroid cancer Diagnosis
(DTC) includes papillary thyroid carcinoma The steps involved in the diagnosis of a solitary
(PTC) and follicular thyroid carcinoma (FTC). nodule are determination of function by labora-
The age-adjusted annual incidence is 0.54 cases tory evaluation, radioiodine scan, and either nee-
per 100,000 children or adolescents, 1 per dle biopsy or open surgical biopsy and excision.
1,000,000 at 10 years of age, 1 per 200,000 from Determination of free T4 and TSH can be helpful;
10 to 14 years of age, and 1 per 75 at 15–19 years. if thyroid hormone is elevated and TSH sup-
Seventy percent of children present with local pressed, there may be a hyperfunctioning nodule.
metastases and 15 % with distant metastases, Evaluation of the appearance by ultrasound scan
usually in the lungs. Distant metastases will not may help determine if there is carcinoma; irregu-
likely appear on standard imaging but are seen on lar borders, heterogeneous densities, and micro-
RAI scans. Recurrence rates up to 30 % are found calcifications are indicators of greater likelihood
as long as 20 years later. of cancer.
Hypothyroidism 119

Fine-needle aspiration under ultrasound guid- Radiation


ance is indicated in suspicious nodules 0.5–1 cm Outcome studies support the use of radioactive
according to some authorities. FNA is useful in ablation (131I using ablative dosage) of remaining
evaluation of lymph nodes since metastases to tissue or metastases after surgery in children;
lymph nodes is common and presurgical knowl- there is a 20–30 % recurrence rate if both modali-
edge of metastases will help surgical planning. ties of therapy are not combined. Decades of
Needle biopsy is safe and has a low level of study suggested the safety of this modality for
false-negative results, but the experience of the children as young as 5 years. However, recent
local pathologists in performing and reading nee- longer-term follow-up data demonstrated an
dle biopsy specimens is important in making the increased risk of secondary carcinoma four to
choice. Even if a needle biopsy suggests a benign five decades later in tissues near the original thy-
or indeterminate diagnosis, if the nodule fails to roid, but not in the thyroid bed. Thus, concern for
shrink on suppressive thyroxine therapy or grows secondary malignancies after successful RAI
over the ensuing months, it may require open therapy arise; studies in children are sparse, and
excisional biopsy as well. In the absence of expe- details are missing in many long-term follow-
rienced clinical help in the procedure of needle ups, so firm conclusions are not available. There
biopsy or in a situation heavily suggestive of a is the confounding factor that having thyroid Ca
malignant diagnosis, open excisional biopsy is predisposes a patient to risk for secondary malig-
best. Only 60 % of samples obtained through nancies which makes interpretation of studies on
fine-needle aspiration in childhood are diagnos- the issues of secondary malignancies after RAI
tic. New molecular biology techniques offer the treatment more difficult. In addition to reproduc-
promise of improved diagnosis and ultimately tive risks, there is a risk of pulmonary fibrosis
improved prediction of prognosis in children (for those with lung metastases). There are also
with thyroid cancer. risks to family members, especially in children at
Measurement of calcitonin in the basal or cal- the home, exposed to radiation-treated children
cium or pentagastrin-stimulated (it is difficult to if they return home in the days after administra-
get pentagastrin at the time of this writing) state tion of RAI therapy before the radiation has
is indicated if there is suspicion of MCT or the passed out of their body.
finding of a first-degree relative with a ret muta- Preparation for RAI treatment includes main-
tion. Evaluation for pheochromocytoma in taining a low iodine diet and avoiding iodine-
affected individuals is important (see Chap. 10). containing soaps or medication (e.g., contrast,
amiodarone) for 1 month. Low urinary iodine
Treatment of Thyroid Carcinoma excretion is a sign that the patient has successfully
diminished or eliminated iodine from their diet.
Surgery The patient may be rendered hypothyroid by
Total or near total thyroidectomy is performed if withdrawing T4 for 4 weeks before the radioactive
papillary or follicular carcinoma is found to min- dose is to be administered so that the thyroid
imize remaining neoplastic tissue. Treatment by gland will avidly take up iodine due to the ele-
a high-volume surgeon (more than 30 thyroid Ca vated TSH caused by the cessation of T4 replace-
surgeries per year) is important due to the high ment. Alternatively switching to T3 replacement
complication rate with such a diagnosis; even an for the 4 weeks before RAI treatment and, because
experienced surgeon experiences a 15 % compli- of the short half-life of T3, stopping the T3 just the
cation rate including recurrent laryngeal nerve day before treatment will allow patients to elimi-
damage, hypoparathyroidism, hemorrhage, and nate the symptoms of hypothyroidism that would
infection, and children under 6 years are most have occurred if 4 weeks passed without any thy-
susceptible to complications. Surgery is usually roid hormone. This method of using T3 prior to
the course of treatment for hyperfunctioning nod- treatment will not suppress the thyroid tissue’s
ules as they rarely regress spontaneously. avidity for iodine uptake for successful treatment.
120 6 Disorders of the Thyroid Gland

The patient must not be constipated or retained suppurative thyroiditis. The viral condition is
iodine will affect the GI tract. At the time of RAI accompanied by a lymphocytosis on CBC, and
administration recombinant human TSH (rTSH) the bacterial form, by an elevated WBC count,
may be invoked to further ensure that the thyroid with a “shift to the left.” Bacterial infections are
takes at the radioactive iodine, but this treatment treated with appropriate antibiotics, and viral
is not FDA approved for children and is extremely infections, with anti-inflammatory medications
expensive. until inflammation resolves.
Suppression of remaining tissue by replace-
ment therapy with thyroxine administration is Hyperfunctioning Nodule
carried out thereafter. This raises the risk of Hyperfunctioning nodules may lead to symptoms
effects on behavior and growth as suppressive of hyperthyroidism. They may be solitary or mul-
doses of thyroxine are higher than replacement tiple. A RAI scan will demonstrate one or more
dose. Serum thyroglobulin determination will “hot” nodules that take up tracers, while the rest
indicate if there is any remaining thyroid tissue of the thyroid is “cold” and does not. Due to the
(normal or neoplastic) but cannot be performed if concern that the hot thyroid tissue will take up
there are thyroglobulin antibodies present. If thy- high levels of radioactivity but the cold regions
roglobulin antibodies exist, they may be mea- will have lower levels and the concern of the
sured at intervals as reduction of thyroid/tumor effect of nonablative radioactivity exposure in the
tissue will lower the antibody level. Follow-up cold tissue in younger children, a surgical
highly by sensitive US scans are preferred over approach is usually preferred.
RAI scans to minimize further radiation exposure.
Recombinant human TSH may be used to stimu-
late the remaining thyroid tissue before radioac- Further Reading
tive iodine scans are performed. Treatment of
metastases that take up iodine may be accom- 1. Rivkees SA. Chapter 12—thyroid disorders in chil-
dren and adolescents. In: Sperling MA, editor.
plished by large doses of radioiodine.
Pediatric endocrinology. Philadelphia, PA: Elsevier;
2014. 4th ed; 2014. p. 444–470.
2. Vliet GV, Deladoëy J. Chapter 7—disorders of the
Painful Thyroid Glands thyroid in the newborn and infant. In: Sperling MA,
editor. Pediatric endocrinology. Philadelphia, PA:
Elsevier; 2014. 4th ed; 2014. p. 186–208.
Tenderness of the thyroid gland may indicate a 3. Leger J, Gelwane G, Kaguelidou F, Benmerad M,
viral infection (subacute thyroiditis) or a bacte- Alberti C. Positive impact of long-term antithyroid
rial infection (suppurative thyroiditis). Either of drug treatment on the outcome of children with
Graves’ disease: national long-term cohort study.
these conditions will be accompanied by an ele-
J Clin Endocrinol Metab. 2012;97(1):110–9.
vated erythrocyte sedimentation rate. Subacute 4. Bahn RS, Burch HB, Cooper DS, Garber JR,
thyroiditis also called de Quervain thyroiditis Greenlee MC, Klein I, Laurberg P, McDougall IR,
may follow a viral illness such as mumps. The Montori VM, Rivkees SA, Ross DS, Sosa JA, Stan
MN. Hyperthyroidism and other causes of thyro-
course usually includes a phase of hyperthyroid-
toxicosis: management guidelines of the American
ism due to release of preformed thyroid hormone; Thyroid Association and American Association of
during this phase the RAI uptake is decreased, Clinical Endocrinologists. Endocr Pract. 2011;
due to damage to the thyroid gland. The course of 17(3):456–520.
5. De Vries L, Bulvik S, Phillip M. Chronic autoimmune
the viral illness may last 6–9 months and then
thyroiditis in children and adolescents: at presentation
spontaneously improve. Suppurative thyroiditis and during long-term follow-up. Arch Dis Child.
might be accompanied by fever, sore throat, 2009;94(1):33–7.
hoarseness, or dysphagia, most of which are 6. American Academy of Pediatrics, Rose SR, Section
on Endocrinology and Committee on Genetics,
absent in subacute disease. It is important to
American Thyroid Association, Brown RS, Public
search for a pyriform sinus fistula by barium Health Committee, Lawson Wilkins Pediatric
swallow, which may play a role in the etiology of Endocrine Society, Foley T, Kaplowitz PB, Kaye CI,
Further Reading 121

Sundararajan S, Varma SK. Update of newborn 16. Fu J, Dumitrescu AM. Inherited defects in thyroid
screening and therapy for congenital hypothyroidism. hormone cell-membrane transport and metabolism.
Pediatrics. 2006;117(6):2290–2303. Best Pract Res Clin Endocrinol Metab. 2014;
7. Rivkees SA, Mazzaferri EL, Verburg FA, Reiners C, 28(2):189–201.
Luster M, Breuer CK, Dinauer CA, Udelsman R. The 17. Negro R, Stagnaro-Green A. Diagnosis and manage-
treatment of differentiated thyroid cancer in children: ment of subclinical hypothyroidism in pregnancy.
emphasis on surgical approach and radioactive iodine BMJ. 2014;349:g4929.
therapy. Endocr Rev. 2011;32(6):798–826. 18. Luongo C, Trivisano L, Alfano F, Salvatore D. Type 3
8. LaFranchi SH. Approach to the diagnosis and treat- deiodinase and consumptive hypothyroidism: a com-
ment of neonatal hypothyroidism. J Clin Endocrinol mon mechanism for a rare disease. Front Endocrinol.
Metab. 2011;96(10):2959–67. 2013;4:115.
9. Ferrara AM, Onigata K, Ercan O, Woodhead H, Weiss 19. Ortiga-Carvalho TM, Sidhaye AR, Wondisford
RE, Refetoff S. Homozygous thyroid hormone recep- FE. Thyroid hormone receptors and resistance to thy-
tor beta-gene mutations in resistance to thyroid hor- roid hormone disorders. Nat Rev Endocrinol. 2014;
mone: three new cases and review of the literature. 10(10):582–91.
J Clin Endocrinol Metab. 2012;97(4):1328–36. 20. Francis GL, Waguespack SG, Bauer AJ, Angelos P,
10. Rivkees SA. Pediatric Graves’ disease: management Benvenga S, Cerutti JM, Dinauer CA, Hamilton J,
in the post-propylthiouracil Era. Int J Pediatr Hay ID, Luster M, Parisi MT, Rachmiel M, Thompson
Endocrinol. 2014;2014(1):10. GB, Yamashita S; American Thyroid Association
11. Glaser NS, Styne DM. Predicting the likelihood of Guidelines Task Force. Management Guidelines for
remission in children with Graves’ disease: a pro- Children with Thyroid Nodules and Differentiated
spective, multicenter study. Pediatrics. 2008;121(3): Thyroid Cancer. Thyroid. 2015;25(7):716–59.
e481–8. 21. Marti JL, Jain KS, Morris LG. Increased risk of sec-
12. Zimmerman D. Thyroid carcinoma in children and ond primary malignancy in pediatric and young adult
adolescents: diagnostic implications of analysis of patients treated with radioactive iodine for differenti-
the tumor genome. Curr Opin Pediatr. 2013;25(4): ated thyroid cancer. Thyroid. 2015;25(6):681–7.
528–31. 22. LaFranchi S. Acquired hypothyroidism in childhood
13. Rivkees SA. Pediatric Graves’ disease: controversies and adolescence. UpToDate,http://www.uptodate.
in management. Horm Res Paediatr. 2010;74(5): com/contents/acquired-hypothyroidism-in-childhood-
305–11. and-adolescence
14. Cunniff C, Frias JL, Kaye C, Moeschler JB, Panny SR, 23. LaFranchi S. Clinical features and detection of con-
Trotter TL, Comm G. Health supervision for children genital hypothyroidism. http://www.uptodate.com/
with Down syndrome. Pediatrics. 2001;107(2):442–9. contents/clinical-features-and-detection-of-
15. Loomba-Albrecht LA, Bremer AA, Styne DM, congenital-hypothyroidism
Glaser NS. High frequency of cardiac and behav- 24. LaFranchi S. Treatment and prognosis of congenital
ioral complaints as presenting symptoms of hyper- hypothyroidism. http://www.uptodate.com/contents/
thyroidism in children. J Pediatr Endocrinol Metab. treatment-and-prognosis-of-congenital-hypothyroidism
2011;24(3-4):209–13.
Disorders of Calcium Metabolism
and Bone 7

Calcium is essential for skeletal integrity and serum of albumin under 4 g/dL. Ionized calcium
plays an essential role in the function of all of the is affected by pH as alkalosis increases protein
living cells of the body. The intracellular and binding limiting ionized calcium which may pre-
intravascular concentration of calcium in blood is cipitate symptoms of hypocalcemia and acidosis
maintained within a narrow range. Increases or releases calcium from protein binding raising
decreases in serum and intracellular calcium ionized calcium. Calcium also varies by age
concentrations may be caused by a wide range of (see Chap. 15). Thus, the physiologic state of
etiologies and can lead to profound changes in the individual must be considered when inter-
organ function. preting either total or ionized calcium.
There are several ions and hormones that
control calcium. Thus, parathyroid hormone,
Calcium Metabolism 1,25-dihydroxyvitamin D, and, more likely in
pathologic situations, calcitonin and parathyroid
Storage and Absorption of Calcium hormone-related peptide (PTHRP) affect serum
calcium. Plasma phosphate and magnesium also
The majority of the body’s store of calcium resides play an important role in aspects of the regulation
in the skeleton, with about 1 % in the recently of calcium.
deposited bone, a pool that can mobilize more eas- Calcium is taken up from the gastrointestinal
ily, and only 1 % of the body’s calcium is found in tract by two mechanisms. The paracellular mecha-
the serum. Calcium is measured as total calcium, nism occurs when calcium is taken up through the
comprising protein bound and the ionized form. tight junctions between cells and is facilitated by
Ionized calcium, the form that causes physiologic proteins including claudins. Active transport of
effects, comprises about 45 % of the total serum calcium across the gastrointestinal cells from the
calcium; ionized calcium measurements are now lumen is the major process involved in calcium
readily available. Ionized calcium is tightly regu- absorption, and this process can be influenced by
lated within a narrow range of values. Of the 1,25-dihydroxyvitamin D; inability to respond to
remainder, 50 % circulates bound to albumin and 1,25-dihydroxyvitamin D leads to hereditary
globulin, and the rest is associated with other ions. 1α,25(OH)2D-resistant rickets (HVDRR).
Thus, total calcium must be interpreted in light of The gastrointestinal (GI) tract can increase the
the amount of circulating protein, because in states efficiency of calcium absorption by active trans-
of low protein total calcium decreases; correction port in states of diminished calcium intake
may be calculated as total serum calcium level through vitamin D-dependent process. However if
declines by 0.8 mg/dL for every 1 g/dL decrease in oral intake is less than 200 mg/day, excretion

© Springer International Publishing Switzerland 2016 123


D.M. Styne, Pediatric Endocrinology, DOI 10.1007/978-3-319-18371-8_7
124 7 Disorders of Calcium Metabolism and Bone

from stool exceeds the amount of absorption, as a result. As a reflection of the central role of
leading to a net loss of calcium which cannot be calcium in cardiac cell function, hypocalcemia
remedied by the GI tract. On the other hand the can lead to prolongation of the QT interval on an
GI tract limits active calcium absorption if GI electrocardiograph.
intake increases above 1,000 mg/day, but passive
absorption cannot be limited; thus excessive oral
calcium intake can cause hypercalcemia (e.g., the The Calcium-Sensing Receptor
milk alkali syndrome). Endocrine factors affect-
ing calcium absorption include growth hormone Maintenance of serum calcium is accomplished
and estradiol which increase absorption and thy- by changes triggered by the calcium-sensing
roid hormone and glucocorticoid which decrease receptor (CASR (+601199 CALCIUM-SENSING
absorption. RECEPTOR; CASR)), a plasma membrane
About 95 % of calcium that enters the renal G-protein-coupled receptor that is expressed in
tubule is reabsorbed. Calcium is resorbed in the the parathyroid hormone-producing chief cells of
renal tubules mainly by passive paracellular pro- the parathyroid gland where parathyroid hormone
cesses generally similar to those in the gastroin- is produced and released, the cells lining the kid-
testinal tract although active transcellular ney tubule where calcium is resorbed and the C
resorption occurs as well under the control of cells of the thyroid gland where calcitonin is pro-
parathyroid hormone. Renal tubular resorption is duced and released. The CASR can sense small
affected by dietary intake, physiologic status, and changes in circulating calcium concentrations,
various genetic conditions. Resorption of cal- and genetic alterations in the receptor are respon-
cium is stimulated by parathyroid hormone and sible for normal variation in serum calcium
parathyroid hormone-related protein, but gluco- ranges as well as several diseases as described
corticoids and mineralocorticoids decrease below. The CASR has a “set point” which is the
resorption. Increased excretion occurs in states of calcium concentration at which PTH secretion is
increased calcium intake, hypercalcemia (in the half-maximal; if the set point is too high, hyper-
absence of hypercalciuric hypercalcemia), loop calcemia will result, and if the set point is too
diuretics, and increased plasma volume. low, hypocalcemia may occur. In the normal state
Serum calcium is regulated within close limits an increase in serum calcium will stimulate the
by several hormones that balance GI intake with CASR to suppress parathyroid hormone secre-
urinary or fecal loss; the skeleton serves as a reser- tion and to decrease the resorption of calcium in
voir of calcium, so that calcium may be mobilized the renal tubule. However, mutations can cause
when needed or stored if necessary. In disorders of an increase or decrease in sensitivity of CaSR
calcium metabolism, excessive calcium may be leading to hypo- and hypercalcemia respectively.
removed from the bone, or, alternatively, defi- An example is familial hypocalciuric hypercalce-
ciency in various factors may impair bone forma- mia in which a decrease of sensitivity of the
tion, leading to one of several bone diseases. calcium-sensing receptor leads to hypercalcemia
Besides increasing bone strength, calcium and retention of calcium in the urine. Other tis-
plays a major role in cellular function by acting sues express CASR demonstrating a remarkable
as an intracellular second messenger as described range of effects changes in serum calcium may
in the beta cell, cardiac cells, and other cells else- bring about.
where in this book. Calcium plays an essential
role in the secretion of neurotransmitters, nerve
conduction, and muscle function. Hypocalcemia Phosphate
leads to increased neuromuscular excitability
leading to tremor; paresthesia around the mouth, Phosphorus in the form of phosphate is another
fingers, and toes; muscle cramps; tetany; laryngo- major component of bone but also plays an
spasm; stridor or seizures; and apnea in neonates important role in cell function throughout the
Calcium Metabolism 125

body involved in such processes as membrane (or calcitriol) and decreases PTH secretion.
receptor activation and in the formation of high- Secretion of FGF23 is normally inhibited by
energy compounds. About 80 % of phosphate is hypophosphatemia. FGF23 is coded by the
found in bone, 9 % in skeletal muscle, 10 % in phosphate regulating endopeptidase homolog,
the viscera, and only about 0.5 % in extracellular X-linked, gene (PHEX). Hypophosphatemic
fluid in phospholipids, phosphate esters, and free rickets is characterized by elevations of FGF23
inorganic phosphate, the latter being the common due to inactivating mutations in PHEX. New
measurement. Serum phosphate varies with age clinical research studies are attempting to treat
far more than serum calcium; for example, con- the condition by utilizing antibodies to FGF23.
centrations in premature infants are above FGF23 can be measured in commercial laborato-
6.4 mg/dL, in children are 4.2–6.4 mg/dL, and in ries; the assays are more accurate in elevated
older teenagers are 2.2–4.3 mg/dL. states than reduced states of FGF23. FGF23 is
Phosphate is absorbed in the small bowel also elevated in the tumoral calcinosis syndrome
mainly by active processes directly stimulated by which is likely a cause of the abnormal phosphate
1,25-dihydroxyvitamin D and therefore indi- metabolism in that condition.
rectly stimulated by hypocalcemia and parathy- Phosphate can shift from extracellular to intra-
roid hormone. In states of phosphate deficiency a cellular locations in many conditions, and this
low phosphate level stimulates the production of may present as hypophosphatemia. Insulin treat-
more 1,25-dihydroxyvitamin D and thereby ment of diabetic ketoacidosis is one example of
increases phosphate absorption. this phenomenon and is also found in the refeed-
Phosphorus with calcium forms the hydroxy- ing syndrome, e.g., after treatment of anorexia
apatite molecules (Ca10(PO4)10(OH)2) which are nervosa and hungry bones syndrome noted below.
responsible for the strength of bone and make up Respiratory alkalosis is also associated with intra-
85 % of skeletal weight. As serum calcium rises cellular shift of phosphate.
phosphate decreases and vice versa; the product of Since phosphate is mainly an intracellular ion,
calcium times phosphorus in a normal state is 30. evaluation of the true state of phosphate abun-
If the product rises above 30, ectopic calcification dance is difficult due to variation throughout the
may occur. Phosphate is absorbed in a process day in serum samples. Tubular reabsorption of
similar to calcium with both active and passive phosphate can be carried out on a 3-h timed urine
transport. About 30 % of phosphate absorption is collection for phosphate and creatinine matched
passive. Phosphate is widely available in food, with serum values of phosphate and creatinine
and passive absorption is proportional to the after an 8-h fast. The percent tubular reabsorption
amount of intake. Active intestinal phosphate of phosphate is calculated, and if it is high in
absorption is increased by 1,25-dihydroxy- times of hyperphosphatemia or low in times of
vitamin D. The kidney is the organ that is mostly hypophosphatemia, it indicates a disorder of the
responsible for the regulation of phosphate. proximal tubule. Excessive FGF23 will cause low
Phosphorus is resorbed in the kidney tubules by an 1,25-dihydroxyvitamin D and low percentage
active process which is increased by hypophos- reabsorption of phosphate, while diminished
phatemia but decreased by hyperphosphatemia FGF23 will cause increase in the reabsorption of
and parathyroid hormone and parathyroid hor- phosphate and 1,25-dihydroxyvitamin D.
mone-related protein. A hallmark of the hypo-
parathyroid state is elevated serum phosphate due
to increased resorption. Magnesium
Fibroblast growth factor 23 (FGF23) is a
member of the phosphatonin family which Magnesium is necessary in adequate quantities to
decreases phosphate resorption in the renal tubule allow the secretion of parathyroid hormone and
and inhibits 1-α-hydroxylase, thereby decreasing for its interaction with the parathyroid hormone
the formation of 1,25-dihydroxyvitamin D receptor. Only about 1 % of the body’s magnesium
126 7 Disorders of Calcium Metabolism and Bone

stores are extracellular. About 30 % of magnesium measures the various inactive forms of PTH that
in the circulation is protein bound with the rest have a longer half-life. Unfortunately because
being free of protein. Thus, hypomagnesemia is a parathyroid hormone is secreted intact and as the
cause of the hypoparathyroid state and hypocal- inactive carboxy terminus of PTH as well, assays
cemia; if this is the etiology of hypocalcemia, must be highly specific to measure the active
repletion of magnesium will cure it. The absorp- parathyroid hormone in the circulation. In fact
tion of magnesium in the gastrointestinal tract the lack of specificity of various assays leads to
occurs generally by passive mechanisms although significant differences in values, especially in the
there is an active component which if altered by presence of renal disease where the terminal mol-
loss of function of the transporter can lead to sec- ecules are present in higher concentrations; it is
ondary hypocalcemia. The active resorption of important to know the characteristics of the assay
magnesium in the renal tubule is stimulated by being used and what it actually measures.
PTH and inhibited by hypermagnesemia and The CSAR stimulates increased production
hypercalcemia. When magnesium stores are low, and secretion of PTH when it senses a low calcium
renal resorption increases. Hypomagnesemia concentration and inhibits the process when cal-
may occur because of lack of absorption or due to cium concentration is high. High serum phosphate
renal leaks of magnesium. Some causes of sec- enhances secretion of PTH, while high 1,25-dihy-
ondary hypomagnesemia are Bartter syndrome, droxyvitamin D decreases secretion of PTH.
renal tubular acidosis, acute renal failure, chronic Parathyroid hormone interacts with its seven-
inflammatory bowel disease, or postsurgical transmembrane domain G-protein-coupled type 1
intestinal resection. Evaluating a spot magne- receptor, type 1 PTH/PTHrP which is found in the
sium-to-creatine ratio in urine will determine kidney and on osteoblasts but also on other tissues.
whether it is due to a renal leak if the ratio is high Parathyroid hormone interacts with its receptor to
or magnesium depletion if the ratio is low. stimulate adenyl cyclase, and measurement of
Hypermagnesemia will also suppress parathy- nephrogenic cAMP (by measuring plasma and uri-
roid hormone secretion. nary cAMP) is a measure of parathyroid hormone
activity; in some types of pseudohypoparathyroid-
ism nephrogenic cAMP does not rise after para-
Parathyroid Hormone thyroid hormone administration. The effects of
and Parathyroid Hormone-Related parathyroid hormone or parathyroid hormone-
Protein related protein are inhibiting renal resorption of
phosphate in the proximal tubule of the nephron,
Parathyroid hormone (PTH) is a polypeptide pro- increasing renal resorption of calcium in the distal
duced in the chief cells of the four parathyroid tubule, increasing renal 1α-hydroxylation of
glands formed by the third and fourth pharyngeal 25-hydroxyvitamin D which in turn increases
arches; a fifth parathyroid gland may be located intestinal calcium absorption, and increasing the
within the thyroid gland in some individuals (but intestinal absorption of calcium. PTH mobilizes
some people have as many as seven). calcium and phosphorous from the skeleton by
PTH is synthesized as a larger prehormone simulating differentiation of osteoclasts, the cells
that is metabolized to the mature 1–84 parathy- that cause bone resorption, from circulating mono-
roid molecule. The active portion of parathyroid cytic hemopoietic cells, stimulating bone remodel-
hormone is in the first 34 amino acids in the N or ing, and releasing calcium and phosphorus
amino terminal region. Soon after secretion the hydroxyapatite in the bone.
parathyroid hormone molecule is cleaved into Parathyroid hormone-related protein
C-terminal and N-terminal moieties. There are (PTHrP) while important in fetal calcium
various assays for PTH; the amino terminus metabolism is produced after birth by various
assay measures the biologically active form that tumors and may serve as a tumor marker for
has a shorter half-life, while the C terminal assay these conditions. It is not regulated by a
Calcium Metabolism 127

feedback inhibition as is parathyroid hormone chylomicrons which are delivered to the liver. In
and therefore is continuously produced. PTHrP the liver 25(OH)vitamin D is synthesized by
is formed locally in many tissues and acts in a 25-hydroxylation of either form of vitamin D.
paracrine manner on nearby tissue, in contrast to 25(OH)vitamin D values reflect the body’s stores
the endocrine action of PTH which acts at a dis- of vitamin D, and measurements are available
tance from the parathyroid gland; however if clinically. 25(OH)vitamin D is transported to the
PTHrP is produced in great enough quantities, it glomerulus bound to vitamin D-binding protein
also acts as an endocrine substance which raises where it is filtered, resorbed, and in the mitochon-
serum calcium. PTHrP is responsible for the dria undergoes 1α-hydroxylation to form
hypercalcemia of malignancy. 1,25-dihydroxyvitamin D (or calcitriol), the active
factor in vitamin physiology and the molecule
which can interact with the vitamin D receptor.
Vitamin D and Its Metabolites (Fig. 7.1) Measurement of 1,25-dihydroxyvitamin D does
not reflect body stores and is not as useful as the
Vitamin D is responsible for the absorption of measurement of 25(OH)vitamin D with the excep-
calcium and phosphorus from the GI tract and tion of certain metabolic conditions such as loss
assists in the mineralization of the skeleton by pro- of function of 1-α-hydroxylase. 1-α-Hydroxylase
viding calcium and phosphorus to the osteoblasts. is increased if serum Ca and phosphate are low,
Endogenous vitamin D3 (or cholecalciferol) thereby increasing the active vitamin D moiety,
can be considered a steroid hormone, which is and is also increased by parathyroid hormone
produced from 7-dehydrocholesterol, a choles- (PTH), PTH-related protein (PTHrP), calcitonin,
terol metabolite, by the effect of sunlight on the growth hormone (GH), prolactin, and insulin-like
skin, which breaks the beta ring of the cholesterol growth factor (IGF)-1. Increased serum Ca and
molecule, followed by a rearrangement of the phosphorous and 1,25-dihydroxyvitamin D all
molecule into the characteristic form of cholecal- decrease the activity of 1-hydroxylase in a nega-
ciferol caused by the heat of the body. The pro- tive-feedback loop. Calcitriol also decreases PTH
duction of cholecalciferol proceeds faster with a synthesis and secretion by negative feedback.
lighter complexion than in dark-skinned individu- Calcitriol is inactivated in the liver and kidney
als so that 10–15 min of midday sun exposure will and other organs with increased inactivation in
be adequate for light-skinned individuals, two to hypercalcemia and hyperphosphatemia and with
three times more for dark-skinned individuals and increased FGF23. Calcitriol formation is increased
six to ten times more for very dark-skinned indi- in several granulomatous conditions as noted
viduals to produce adequate vitamin D; Winter below under other causes of hypercalcemia.
sun exposure would be more. The recommenda- Vitamin D deficiency can occur with absence
tion to limit sun exposure through the use of sun- of adequate vitamin D intake. However, sun
screen or sun avoidance will not allow these exposure will increase the production of vitamin
periods of exposure; vitamin D deficiency may D and will mitigate the situation. Cloudy days or
develop without additional supplemental oral sun exposure through a window will not allow
vitamin D intake. While vitamin D might be found the skin to produce much vitamin D. However in
in oily fish, a customary healthy diet does not con- the present era of concern over skin cancer due to
tain a sufficient amount of vitamin D, and thus sun exposure in children and the recommenda-
supplementation with vitamin D is necessary fail- tions for the use of sunscreen, sun exposure in
ing adequate sun exposure. Oral vitamin D is gen- children must necessarily be limited so that oral
erally found in the form of vitamin D2 or vitamin D intake is usually essential. The NIH
ergocalciferol derived from plants, while D3 or does not recommend sun exposure for more than
cholecalciferol is derived from animal sources. a few minutes without protection as a manner of
They both follow the same metabolic pathways. increasing vitamin D.
Vitamin D is absorbed with other fats since it is a Vitamin D deficiency can occur with various
fat-soluble vitamin and is incorporated into gastrointestinal disorders including celiac disease
128 7 Disorders of Calcium Metabolism and Bone

HO
7-Dehydrocholesterol (skin)

Sun + –
light UV Sun avoidance
CH2
HO
Vitamin D3

Liver – Liver disease


25-Hydroxylase Anticonvulsants
Biliary disease

OH

CH2

25-Hydroxyvitamin D

1α -Hydroxylase Aluminum?? Lead??


Low Ca++ –
Renal disease
Low P
+ PTH Vitamin D - dependency rickets
X-linked hypophosphatic rickets
Kidney

OH + Gut
Ca++
+ Ca++
CH2 Maintain normal
blood calcium
HO OH
1,25-Dihydroxyvitamin D3 Ca– Ca++
– Aluminum
+
Ca++
Calcitonin Mineralization
Demineralization
PTH
1,25(OH)2D

Fig. 7.1 The pathway for the regulation of serum cal- vitamin D is 25-hydroxylated in the liver. An additional
cium and the biosynthesis of 1,25(OH)2 vitamin D 1-hydroxylase step occurs in the kidney to produce the
(redrawn from a figure by Chesney R, in Styne DM, ed. active moiety 1,25-dihydroxyvitamin D. The location of
Pediatric endocrinology: House officer series.). The parathyroid hormone and vitamin D effects is indicated
child exposed to adequate sunlight will convert 7-hydroxy along with factors which may inhibit appropriate effects.
cholesterol to vitamin D3. With normal liver function and Redrawn from an original figure from R. Chesney with
no exposure to medications that may inhibit the process, permission

short bowel syndrome, biliary obstruction, and although calcitriol will be necessary in other con-
any cause of fat malabsorption. ditions in which endogenous conversion to
Cholecalciferol and ergocalciferol are fat 1,25-dihydroxyvitamin D is limited.
soluble, so an overdose will exert effects for In severe vitamin D deficiency when treat-
months as it is released from fat. Calcitriol is not ment is offered, the patient may experience hypo-
fat soluble, so daily dosing or several doses per calcemia due to the hungry bone syndrome. Thus,
day are needed and its effects last only a short the bones long deprived of calcium will with
time. It is easier, and cheaper, to treat with chole- vitamin D treatment avidly take up calcium lead-
calciferol than calcitriol in vitamin D deficiency ing to lower serum calcium levels. Thus, calcium
Calcium Metabolism 129

augmentation is necessary. There may be associ- vitamin D, limiting the production of 1,25(OH)
ated hypophosphatemia, hypomagnesemia, and vitamin D, and both limit further increase in cal-
hyperkalemia as described below. cium. Further, a decrease in serum calcium trig-
Vitamin D is also ingested from various vita- gers a release of PTH, which increases 1,25(OH)
min D-supplemented dairy products (D2 or ergo- vitamin D production, and both of these factors
calciferol is the most common form in food increase serum calcium values. Vitamin D recep-
although D3, cholecalciferol, is also added to tors in the parathyroid hormone cells and calcitriol
food) or other foods that may already contain can suppress parathyroid hormone secretion and
vitamin D (e.g., oily fish such as salmon and cod also suppresses calcitonin secretions.
liver oil). The fetus is completely dependent upon the
1,25-Dihydroxyvitamin D interacts with the maternal supply of vitamin D. 25-Hydroxyvitamin
nuclear transcription factor, the vitamin D recep- D crosses the placenta, but if the mother is defi-
tor (VDR), which is a member of the steroid cient, the infant will be as well.
receptor superfamily. The complex of the vitamin
D molecule and its receptor forms a heterodimer
with the retinoid receptor and binds to a vitamin Calcitonin
D-responsive element in the DNA on a specific
gene which is responsive to vitamin D such as Calcitonin is a polypeptide hormone produced by
genes for calcium binding protein or the parafollicular or C cells of the thyroid gland.
24-hydroxylase. VDR is expressed throughout Calcitonin decreases serum calcium by inhibiting
the body but for the purposes of this discussion is the action of osteoclasts on bone and by restoring
found in the GI tract, the renal tubule, and the calcium into bone, thus inhibiting the effect of
osteoblasts. The vitamin D receptor can reduce PTH on bone. Increased calcitonin is secreted at
expression of 1α-hydroxylase while increasing times of hypercalcemia by signaling through the
CYP24 which inactivates 1,25-dihydroxy- CASR, and calcitonin secretion is decreased at
vitamin D. Increased expression of the gene for times of hypocalcemia. Calcitonin is a tumor
the VDR occurs with exposure to 1,25-dihydroxy- marker for medullary carcinoma of the thyroid, a
vitamin D, while gene expression is decreased by condition for which it is secreted in excess
glucocorticoids. 1,25-Dihydroxyvitamin D (see Chap. 10). Remarkably in spite of the biologic
increases absorption of calcium and phosphorous activities detailed above, absence of calcitonin or
from the small intestine and assists PTH in the increased endogenous levels of calcitonin do not
resorption of bone to increase serum calcium and significantly affect serum calcium levels.
by the production of osteoclasts. 1,25-Dihydroxy- Nonetheless, exogenous calcitonin is used as
vitamin D also assists in the mineralization of the therapy for hypercalcemia although this is being
skeleton by maintaining serum Ca and PO4 within replaced by bisphosphonates in some situations.
appropriate ranges to allow deposition of
hydroxyapatite, the calcium–phosphorus com-
pound which comprises the mineral portion of Alkaline Phosphatase
bone. In addition, 1,25-dihydroxyvitamin D
increases retention of calcium from the renal Alkaline phosphatases are a family of metalloen-
tubule to decrease the loss of calcium in the urine, zymes responsible for catalyzing the hydrolysis
while it decreases phosphorus retention from the of phosphate esters and are found in the intesti-
nephron. All forms of vitamin D circulate bound nal, placental, and liver isoforms. Post-translation
to a vitamin D-binding protein of 52 kDa. modification leads to tissue-specific forms found
A complex feedback loop is found in this sys- in bone, liver, and kidney which can be separated
tem of calcium regulation, as an increase in due to their different isoelectric points and heat
serum calcium decreases PTH secretion, which stability by a clinical laboratory. The bone form
in turn decreases the 1α-hydroxylation of 25(OH) is secreted by osteoblasts and increases with min-
130 7 Disorders of Calcium Metabolism and Bone

eralization of bones. Total alkaline phosphatase Hypocalcemia in the Infant


measurements characteristically reflect the liver
and bone forms. The alkaline phosphatase from Normal term infants have serum Ca greater than
bone rises during childhood growth and roughly 8.5 mg/dL and ionized Ca more than 4.5 mg/dL.
relates to growth rate. But bone-derived alkaline The normal values are controversial in premature
phosphatase will also increase in diseases of bone infants, but as a guide, hypocalcemia may be
turnover such as rickets, fibrous dysplasia defined as a serum Ca below as 7.5 mg/dL and
(including McCune–Albright syndrome), and ionized Ca below 4.4 mg/dL(or 1.1 mmol/L) if
juvenile Paget’s disease reflecting bone activity. birthweight is over 1,500 g and below 7 mg/dL
Low levels of alkaline phosphatase lead to the and ionized Ca below 3.6 mg/dL (0.9 mmol/L) if
severe crippling conditions hypophosphatasia birthweight is under 1,500 g. Symptoms of hypo-
due to mutations in the alkaline phosphatase calcemia are more nonspecific in newborns than
gene (ALPL) (#241500 ICD+HYPOPHOSPHA in older children; poor feeding, emesis, and apnea
TASIA, INFANTILE) which is autosomal reces- may be the only signs. More specific signs of
sive, (#241510 ICD+HYPOPHOSPHATASIA, hypocalcemia that might be found in older infants
CHILDHOOD) which is autosomal recessive, include tachycardia, irritability, jitteriness, tetany,
and (#146300 ICD+HYPOPHOSPHATASIA, laryngospasm, and seizures.
ADULT) which can be autosomal recessive or
autosomal dominant. At the time of this writing Early Neonatal Hypocalcemia
new therapy to replace enzymatic activity is For the first few days after birth, the infant will
pending FDA approval for the severe forms. have difficulty regulating serum calcium. During
Markers of bone formation include total and this period just after the infant separates from the
bone-specific alkaline phosphatase, osteocalcin, maternal calcium supply and the maternal regula-
and serum type 1 procollagen. Markers of bone tion of serum calcium, PTH secretion is often
resorption are urinary hydroxyproline, urinary total sluggish, so that transient and mild hypocalcemia
pyridinoline (PYD), urinary free deoxypyridino- and hyperphosphatemia may result. If the mother
line (DPD), urinary collagen type 1 cross-linked herself has hypercalcemia, the fetus will experi-
N-telopeptide (NTX), urinary or serum collagen ence elevated calcium levels and suppress fetal
type 1 cross-linked C-telopeptide (CTX), bone parathyroid hormone secretion; after birth there
sialoprotein (BSP), and tartrate-resistant acid will be a prolonged period of relative hypopara-
phosphatase 5b. thyroidism while the neonatal parathyroid glands
Transient hyperphosphatasemia of infancy recover. Further, an infant has a low glomerular
and early childhood is a benign condition found filtration rate that decreases phosphorus excre-
in children under 5 years of age. Bone and liver tion and causes increased serum phosphorus,
alkaline phosphatase isoforms are elevated which further reduces serum calcium. Transient
although there is no disease in either organ. After episodes of low serum calcium during the first 3
the passage of a few months, alkaline phospha- days after birth is more common in patients who
tase returns to normal. are premature, have intrauterine growth retarda-
tion (IUGR) or are small for gestational age
(SGA), have sepsis, have asphyxiation, or are
Hypocalcemia (Fig. 7.2) infants of diabetic mothers (the worse the control
of blood sugar, the lower the Ca in the baby).
Hypocalcemia can be due to various etiologies This is considered to be early hypocalcemia of
depending upon the age of the child. The signs infancy. Cow’s milk contains more phosphorus
and symptoms of hypocalcemia are likewise than does human milk, even though the calcium
dependent on the age of the child. Thus, we will content is higher: the load of oral phosphorus
consider hypocalcemia in the infant and hypocal- may enhance the tendency to hypocalcemia of
cemia at an older age. infancy to symptomatic levels in infants fed with
Hypocalcemia 131

Fig. 7.2 Flow chart of the Low serum Ca


diagnosis of the etiology of
hypocalcemia. Initial
Serum PO4
differentiation between
high serum phosphate
which would suggest a Low High
disorder of parathyroid
hormone action (light gray
arrows) and low serum Serum Alk phos
phosphate which should be
more likely related to an
High Low
abnormality of renal
function open (dashed
dark lines) or vitamin D Serum PTH
stores or action (solid
black line) is a useful to
approach the problem High Low

25 0 H Vitamin D Magnesium

Normal Low Low Normal

Renal Function Vitamin D Hypo- Hypo-


deficiency magnesemia parathyroidism

Normal Abnormal

Pseudohypo- Renal
parathyroidism Disease

cow’s milk formula. This is less common in due to dietary hyperphosphatemia, which will
breast-fed infants due to the favorable calcium lead to low calcium and high phosphorus; low
and phosphorus ratio. Breast milk is a poor source calcium may trigger PTH secretion and falsely
of vitamin D, especially if the mother has a vita- suggest pseudohypoparathyroidism. Renal dis-
min D-deficient diet, and vitamin D deficiency ease may lead to phosphate retention and elevated
may occur in an infant because of coexistent vita- parathyroid hormone with the clinical presenta-
min D deficiency in the mother; this condition is tion of reduced renal function. Soy formula or the
becoming more common (see below). Thus, early early introduction of cereal into the diet leads to
neonatal hypocalcemia may not occur in breast- an increased intake of phytates which may lead to
feeding infants, but hypocalcemia and rickets hypocalcemia. Hypomagnesemia will decrease
may develop later unless vitamin D supplementa- the secretion of parathyroid hormone leading to
tion of the breast fed neonate is carried out (see hypocalcemia.
vitamin D deficiency below). Other causes of hypocalcemia in this period
Medications given to the newborn that can include hypoparathyroidism. As noted above,
cause hypocalcemia include aminoglycoside the development of parathyroid function takes a
antibiotics, phosphates, citrate, fatty acids, and while after birth, so a transient condition of
bicarbonate. hypoparathyroidism can occur. If hypocalcemia
continues, a search for an etiology for decreased
Late Neonatal Hypocalcemia parathyroid hormone secretion or action is war-
If hypocalcemia occurs after 3 days, it is consid- ranted. If a chest radiograph shows no thymic
ered late neonatal hypocalcemia. This may be shadow or a cardiac defect of the great vessels is
132 7 Disorders of Calcium Metabolism and Bone

found, the diagnosis of DiGeorge syndrome is Diagnosis of Infantile Hypocalcemia


more likely than transient hypocalcemia. The As always differential diagnosis starts with a
DiGeorge syndrome (*188400 DIGEORGE detailed history. The mother’s nutritional status,
SYNDROME; DGS) results from disordered the presence of diabetes, a family history of bone
development of the third and fourth branchial disease, seizures, kidney stones or other indica-
pouches and first to fifth branchial arches tions of disorders of calcium metabolism, com-
derived from neural crest cells. This is usually plications of delivery, and the condition of the
found associated with microdeletions of chro- child in the neonatal period all play a role in the
mosome 22q11.2. This syndrome may be spo- evaluation of the conditions detailed above.
radic or inherited in an autosomal dominant Physical examination will include a search for
pattern and is relatively common with a preva- dysmorphology and disorders of the extremities
lence of 1 in 4,000, similar to congenital hypo- although some of those such as the classic skele-
thyroidism, and is present in 70 % of children tal findings of Albright hereditary osteodystrophy
who appeared to have isolated hypoparathyroid- may not develop until later in life. Diagnostic
ism. The classic DiGeorge syndrome combines studies for hypocalcemia of infancy should be
hypoparathyroidism with abnormalities of the carried before treatment but only if the delay is
aortic arch (such as truncus arteriosus or tetral- judged clinically safe. Thus, obtaining serum
ogy of Fallot) and the thymus (immune defects total and ionized calcium, magnesium, phos-
related to T-cell defects and disordered cellular phate, creatinine, 25 OH vitamin D, 1,25-dihy-
immunity) and impaired cell-mediated immu- droxyvitamin D, PTH (intact), and urinary
nity. Physical findings include hypertelorism, calcium and creatinine concentrations is indi-
short philtrum, micrognathia, malar hypoplasia, cated. If diagnosis has not been clarified measur-
low-set ears, and sometimes cleft palate. ing maternal calcium, phosphorus and PTH level
Velocardiofacial syndrome (#192430 might be useful. Pseudohypoparathyroidism type
VELOCARDIOFACIAL SYNDROME at 1A may present with associated primary congeni-
22q11.2) has the same gene locus as DiGeorge tal hypothyroidism and an elevated TSH. The
syndrome and has, as well, high-arched or sub- combination of hypocalcemia and elevated TSH
mucous cleft palate, retrognathia, short palpe- should point to that possibility. To evaluate the
bral fissures, and VSD. The CATCH-22 possibility of DiGeorge syndrome, CBC and
syndrome is a general term encompassing many CD4 lymphocyte count are performed. A chest
phenotypes caused by mutations at this locus; X-ray to check for a thymic shadow can be done
this syndrome is composed as a cardiac defect, quickly but may not be completely reliable if the
abnormal facies, thymic hypoplasia, cleft pal- infant is severely ill. The ultimate diagnostic tool
ate, and hypocalcemia with deletion at 22q. takes longer, by a fluorescent in situ hybridiza-
Thus, any infant with tetany and hyperphospha- tion (FISH) study for a microdeletion of chromo-
temia should have evaluation for cardiac and some 22q11.2.
immune disease at the least, and a genetic screen
should be considered. The hypocalcemia of Treatment of Newborns
DiGeorge syndrome often resolves after infancy Therapy for asymptomatic hypocalcemia in
only to recur in some cases at times of rapid infants is usually carried out if the calcium in
growth of adolescence. the full-term infant is less than 7 mg/dL or in a
Hypercalciuric hypocalcemia may present in premature infant is less than 6 mg/dL, and treat-
infancy and is discussed below. There are several ment is provided to those who are symptomatic.
variations of loss-of-function mutations in the Mild cases might be treated by a change in for-
PTH1R. Likewise pseudohypoparathyroidism mula to a low phosphorous formula such as PM
may present in an infant and is also discussed 60/40 or one with at least a 4:1 calcium-to-phos-
below. phorus ratio in addition to oral calcium glubionate
Hypocalcemia 133

(30–50 mg/kg/day in four divided doses) or calcium enhanced protein binding of ionized Ca, and this
gluconate (500 mg/kg/day in four to six feed- may precipitate tetany. This situation may occur in
ings per day). If vitamin D or magnesium is low, states of anxiety and is treated by rebreathing CO2
replacement of the deficits is the treatment (classically from a paper bag) to decrease serum
modality. Maintenance values of 1,25-dihydroxy- pH, thereby freeing some calcium from the protein
vitamin D calcium in children with hypopara- into the ionized state in the serum. Signs of long-
thyroidism range from 20 to 40 ng/kg/day. term hypocalcemia include calcification of the
With severe symptoms including seizures and basal ganglia, cataract formation, and poor tooth
laryngospasm, intravenous (i.v.) 10 % calcium enamel formation. Hypomagnesemia will decrease
gluconate (equivalent of 9.3 mg/mL of elemental PTH secretion and can precipitate a temporary
calcium) in a dose of 100 mg/kg or 1 mL/kg phase of hypoparathyroidism, which is treated by
infused over 15 min with a total dose no more remedying the deficit in magnesium rather than by
than 20 mg of elemental calcium per kilogram is administration of vitamin D or calcium.
used. Alternatively, calcium chloride in a dose of
20 mg/kg or 0.2 mL/kg might be substituted with
the same regimen. EKG monitoring for arrhyth- Hypocalcemia in the Child
mias or bradycardia is mandatory at any time i.v. and Adolescent
calcium is administered. If an umbilical venous
catheter is used and the tip is inadvertently is The diagnosis of hypocalcemia after infancy is
inserted in the liver, hepatic necrosis may result also based upon age. Normal values will depend
from IV calcium administration. A repeated dose on the laboratory, but one guideline is as follows:
may be administered after 10–15 min if there has 1–5 years the normal range is 9.4–10.8 mg/dL, at
been no response of the symptoms and every 6 h 6–12 years it is 9.4–10.2 mg/dL, and up to 20 years
but only if clinically indicated for continued it is 8.8–10.2 mg/dL. On occasion asymptomatic
hypocalcemia. Maintenance intravenous calcium hypocalcemia may be uncovered, or mild findings
is 500 mg/kg/day of 10% calcium gluconate. such as muscle soreness or cramps may lead to the
Severe hypomagnesemia as etiology for hypo- diagnosis. More severe hypocalcemia may be
parathyroidism is treated with IM injection or a manifested by three classic physical findings:
1–2-h intravenous infusion of 50 % magnesium
sulfate at a dose of 0.1–0.2 mL/kg, again with 1. The Chvostek sign, in which tapping the facial
careful EKG monitoring. The goal is to achieve a nerve 0.5–1.0 cm below the zygomatic pro-
magnesium level of 1.5 mg/dL. cess and 2 cm anterior to the ear lobe causes a
A decrease in serum ionized Ca2+ leads to the twitch at the angle of the mouth due to con-
signs of hypocalcemia: neuromuscular irritability, traction of the perioral muscles, a demonstra-
causing tetany (heralded by carpopedal spasm, tion of muscular irritability. Almost 30 % of
causing the fingers to extend with ulnar devia- hypocalcemic individuals may not show this
tion), muscle cramps, weakness, lethargy, pares- sign however.
thesias of the extremities, and in the most severe 2. The Trousseau sign, in which a blood pressure
situation, convulsions and/or laryngospasm (stri- cuff is inflated at least 15 mmHg above the
dor or complete closure of the larynx with the risk systolic blood pressure for 2–5 min; a positive
of death) (Table 7.1). Lengthening of the QTc response is indicated by “main d’accoucheur”
interval may be seen on the electrocardiogram position with flexion of the wrist and metacar-
(EKG). Although presentation may appear simi- pophalangeal joints and extension of the inter-
lar to that of a grand mal seizure, in view of the phalangeal joints and adduction of the fingers
tonic/clonic activity, no postictal phase occurs due to carpopedal spasm in the arm to which
with the resolution of hypocalcemia. the cuff was applied. This is reliable in up to
Hyperventilation causing respiratory alkalosis 94 % of cases with hypocalcemia. This can be
will reduce the ionized fraction of calcium by painful however.
134 7 Disorders of Calcium Metabolism and Bone

Table 7.1 Causes of hypocalcemia


Serum Serum Serum Serum Alkaline
Disorder Ca Ca2+ PO4 PTH phosphatase pH Other features
Vitamin D deficiency Low Low Low High High Nl Serum 25 OH
or Nl or Nl vitamin D low,
1,25(OH2) vitamin
D may be normal
Hyperventilation Nl Low Nl Nl Nl High Cured by
rebreathing
Transient hypocalcemia of Low Low Nl/high Low Nl Nl Made worse with
infancy relative high-phosphorus
to Ca feedings
Congenital Low Low High Low Low Nl May be part of
hypoparathyroidism relative DiGeorge syndrome
to Ca
Autoimmune Low Low High Low Low Nl May be part of
hypoparathyroidism relative autoimmune
to Ca polyendocrinopathy
syndrome, type 1
Pseudohypoparathyroidism Low Low High High Low Nl May be associated
relative with Albright
to Ca hereditary
osteodystrophy
Magnesium deficiency Low Low High Low Low Nl May be part of
relative Barter syndrome,
to Ca malabsorption, or
renal disease
Calcium deficiency Low Low Could be Elevated Elevated Nl Rare in present day
low or except for fad diets,
normal vitamin deficiency
with, possibly, sun
avoidance
Renal disease Low Low High High High Possibly Tests of renal
acidotic function are
abnormal
From: Fisher DA, ed. The Quest Diagnostics Manual: Pediatric endocrinology. San Juan Capistrano: Quest Diagnostics
Incorporated 2000.
PTH parathyroid hormone, Nl normal

3. The peroneal sign, in which a tap on the pero- narrowing of the QRS complex, reduced PR
neal nerve near the lateral tibial prominence interval, T wave flattening and inversion and
causes plantar flexion (although tapping any prolonged ST and ST depression.
nerve may lead to contraction of the associ-
ated muscle, other contractions may be more Hypoparathyroidism
difficult to demonstrate). Hypoparathyroidism is a cause of hypocalcemia.
This is partly due to the lack of parathyroid hor-
Clinical symptoms of hypocalcemia in the mone effect on bones in the maintenance of
child include paresthesias, circumoral numb- serum calcium and partly due to the lack of pro-
ness, muscular cramping, tetany, laryngospasm, duction of calcitriol which is dependent on para-
bronchospasm, muscle twitching, carpal–pedal thyroid hormone action.
spasm, and possibly seizures. Lengthening of the While congenital defects of the parathyroid
QTc interval may be seen on the electrocardio- glands are found in the newborn, some of the
gram (EKG) as well as prominent U-wave, conditions described above may occur at an older
Hypocalcemia 135

age. However, most cases of hypoparathyroidism X-LINKED; HYPX at Xq26–27). Other forms
will be due to an autoimmune etiology in child- may be associated with disorders of development
hood. Because adrenal insufficiency due to an of the parathyroid gland associated with renal
autoimmune etiology may accompany autoim- disease and deafness (#146255 HYPOPARA-
mune hypoparathyroidism, the clinician must THYROIDISM, SENSORI-NEURAL DEAFNESS,
have a high index of suspicion for adrenal failure AND RENAL DISEASE; HDR *241400
as fatalities have occurred when it is missed. HYPOPARA-THYROIDISM with sensineural
Autoimmune hypoparathyroidism may occur as deafness). The Sanjad–Sakati (*241410
an isolated case or in association with type 1 HYPOPARA-THYROIDISM-RETARDATION-
polyendocrinopathy syndrome, which also DYSMORPHISM SYNDROME; HRD at
includes chronic mucocutaneous candidiasis and 1q42043) syndrome of fetal and postnatal growth
ectodermal dysplasia (APECED *240300 retardation, seizures, developmental delay, and
AUTOIMMUNE POLYENDOCRINOPATHY dysmorphic facial features is associated with
SYNDROME, TYPE I). Other associated defects autosomal recessive hypoparathyroidism. The
include Addison’s disease, autoimmune hypergo- Kenny–Caffey syndrome (*244460 KENNY-
nadotropic hypogonadism, diabetes mellitus type CAFFEY SYNDROME, TYPE 1; KCS at 1q42–
1, autoimmune disease of the thyroid gland, per- 43) may be inherited in an autosomal dominant,
nicious anemia, vitiligo, alopecia, dental enamel autosomal recessive, or even X-linked pattern
hypoplasia, keratoconjunctivitis, malabsorption, and includes poor growth, stenosis of the bone
chronic active hepatitis, and variable T-cell marrow cavity, and thickened tubular cortical
defects. A defect in the AIRE gene is seen at bones. Hypoparathyroidism might occur with
21q22.3 in APECED (see Chap. 10). The renal dysplasia and deafness (#146255
immunodysregulation, polyendocrinopathy, HYPOPARATHYROIDISM, SENSORI-NEURAL
and enteropathy syndrome (#304790 ICD+ DEAFNESS, AND RENAL DISEASE; HDRA)
IMMUNODYSREGULATION, POLYENDO- in an autosomal dominant pattern in the rare
CRINOPATHY, AND ENTEROPATHY, Barakat syndrome.
X-LINKED; IPEX) is inherited in an X-linked Gain-of-function mutation of the calcium-
pattern and is another autoimmune condition sensing receptor (#601198 HYPOCALCEMIA,
which included hypoparathyroidism in the con- AUTOSOMAL DOMINANT 1; HYPOC1) pres-
stellation of disorders. Chronic inflammation of ents with mild to moderate symptoms of hypocal-
the GI tract with diarrhea, ileus, and villous atro- cemia with hypercalciuria and nephrocalcinosis
phy combines with eczema and atopic conditions and/or nephrolithiasis along with calcification of
of the skin, type 1 diabetes mellitus, autoimmune the basal ganglia. Nephrolithiasis may occur with
hypothyroidism, autoimmune hemolytic anemia calcium oxalate or calcium phosphate kidney
and thrombocytopenia, and other types of stones. Kidney stones form when the product of
immune problems. This condition is caused by an the ions exceeds the solutability factor of the
abnormality of the forkhead box P3 gene ions. Homogeneous nucleation occurs when
(FOXP3, 300292.0001) and usually leads to calcium and oxalate crystals form and then grow
death in infancy or childhood. too large and form calcium oxalate stones.
Nonautoimmune hypoparathyroidism may Heterogeneous nucleation occurs when calcium
occur sporadically or in autosomal dominant and phosphorus combine and cause calcium
or autosomal recessive pattern (#146200 phosphate nuclei in which calcium oxalate is not
HYPOPARATHYROIDISM, FAMILIAL present. Because of the elevated sensitivity of the
ISOLATED; FIH at 3q13) due to a mutation in calcium-sensing receptor in this condition, para-
the PTH gene; patients may have cataracts or thyroid hormone is not detectable or is inappro-
intracerebral calcifications. Hypoparathyroidism priately low. Children with this condition are
in an X-linked pattern may present in a neonate extremely sensitive to vitamin D which in normal
(*307700 HYPOPARATHYROIDISM, doses will cause hypercalcemia. Treatment has
136 7 Disorders of Calcium Metabolism and Bone

been successful with exogenous parathyroid skin, possibly requiring plastic surgical repair.
hormone. The infusion may be repeated if necessary for the
There are other forms of acquired hypopara- relief of symptoms but is followed by 10 % i.v.
thyroidism. Hypoparathyroidism may occur from calcium gluconate at a dose of 500 mg/kg over a
iron deposition of thalassemia (and its therapy) or 24-h period for neonates and 200 mg/kg for
hemochromatosis, copper infiltration from infants or older subjects. Long-term therapy is
Wilson disease. Radiation to the thyroid gland achieved by 1,25(OH)vitamin D (this form elimi-
and parathyroid gland area, trauma, or surgery nates the need for 1-hydroxylation of vitamin D,
also may cause acquired hypoparathyroidism. which is impaired in hypoparathyroidism) given
Surgical hypoparathyroidism may occur after at a dose of 20–40 ng/kg/day (calculated as 0.25–
thyroid surgery. It may be transient because of 0.75 g twice daily), with oral calcium supple-
inflammation of the area, or it may be permanent mentation adequate to maintain serum ionized
because of the removal of parathyroid tissue calcium, if necessary. Urine can be tested for
along with the thyroid tissue. excessive calcium loss by determining urinary
calcium/creatinine ratio (this normally should be
The Diagnosis of Hypoparathyroidism less than 0.2) to evaluate the effects of therapy.
The diagnosis of hypoparathyroidism rests on Renal ultrasound examinations are administered
demonstration of decreased serum ionized cal- at yearly intervals to search for nephrocalcinosis,
cium and increased phosphorus, in the face of which is a complication of vitamin D therapy.
inappropriately decreased serum PTH for the Phosphorus is restricted in the diet until serum
value of serum calcium. Low serum ionized cal- phosphorus decreases to near-normal levels,
cium should, in the normal situation, cause eleva- which should occur when serum ionized calcium
tion of serum PTH, and absence of such a response approaches normal. Serum ionized calcium
suggests inadequacy of PTH secretory ability; should be measured regularly until stability is
complete absence of serum PTH is not necessary achieved to assure that serum calcium remains in
for the diagnosis of hypoparathyroidism. Urinary the normal range. (For a discussion of emergency
calcium excretion is elevated (calcium/creati- treatment of hypocalcemia, see Chap. 14.)
nine ratio greater than 0.2), phosphate excretion
is diminished (tubular reabsorption of phosphate Pseudohypoparathyroidism
is greater than 88 %), and cyclic adenosine Individuals with resistance to PTH have pseudo-
monophosphate (cAMP) is low in the urine of hypoparathyroidism or PHO and have all the
hypoparathyroidism, as PTH normally stimulates features of hypoparathyroidism except that PTH
the production of cAMP in the renal tubule. is elevated, although it is ineffective in control-
Hypoparathyroidism as well as pseudohypopara- ling calcium values due to abnormalities in the
thyroidism causes ectopic calcification, particu- receptor for PTH rendering it unresponsive. This
larly intracranial calcification. condition can occur early in infancy as well as
later in childhood. The phenotype known as
Treatment of Hypoparathyroidism Albright hereditary osteodystrophy (AHO),
Treatment of hypoparathyroidism must first which consists of short stature, obesity, round
address hypocalcemia. For severe symptomatic facies, brachydactyly, and metacarpal or metatar-
hypocalcemia, 10 % calcium gluconate is given sal hypoplasia resulting in shortened forth and
as a dose of 0.5–1 mL/kg infused over 15–20 min fifth fingers and toes, is found in pseudohypo-
up to a total of 10 mL, given with EKG monitor- parathyroidism type 1A (#103580 ICD+PSEUD
ing (bradycardia and asystole are possible com- OHYPOPARATHYROIDISM, TYPE IA;
plications of an increase of calcium). The PHP1A). The condition is due to a loss-of-func-
concentration is decreased to 5% for administra- tion mutation of the Gs-alpha isoform of the
tion in some protocols. During the administration GNAS gene (139320) located at 20q13.32
of i.v. calcium, it is important to avoid extravasa- thereby limiting the effects of parathyroid hor-
tion, which may cause severe sloughing of the mone on its receptor. In an example of parental
Hypocalcemia 137

imprinting, if the mutated gene is inherited from Magnesium Deficiency and Magnesium
the father, AHO results in isolation without Excess
deficient hormone action in a condition known as Magnesium deficiency diminishes PTH secretion
pseudopseudohypoparathyroidism (#612463 as well as PTH-mediated bone resorption. Thus,
ICD+ PSEUDOPSEUDOHYPOPARATHYRO signs of magnesium deficiency are those of hypo-
IDISM; PPHP), whereas if the gene is inherited calcemia which usually accompanies magnesium
from the mother, the child has type 1A PHO as deficiency. EKG changes of hypomagnesemia
described above also with AHO phenotype. are prolonged QTc interval, atrial and ventricu-
Urinary cAMP excretion does not increase after lar ectopy, atrial tachyarrhythmias, and torsades
PTH infusion if the defect is generalized (type de pointes.
1A), and resistance to thyroid-stimulating hor- Magnesium deficiency may be found in the
mone (TSH), antidiuretic hormone (ADH), and newborns who are infants of diabetic mothers or
growth hormone-releasing hormone (GHRH) mothers suffering from eclampsia or have mag-
may occur in conjunction with type IA PHO with nesium deficiency themselves. Hypomagnesemia
the phenotype of AHO. Basal ganglia calcifica- is found in IUGR and prematurity. Nasogastric
tions are common on radiograph, and subcutane- suctioning may deplete the child of magnesium.
ous calcifications occur as well. Other subjects Treatment with loop diuretics, thiazide diuretics,
may have renal unresponsiveness to PTH, but various types of antibiotics, gastrointestinal treat-
responsiveness of the skeleton to PTH, leading to ments, digitalis, and various anticancer medica-
the hyperparathyroid bone disease, osteitis tions (cisplatin, aminoglycosides) can diminish
fibrosa cystica. These subjects have type 1B urinary magnesium reabsorption and result in
pseudohypoparathyroidism (#603233 PSEUDO hypomagnesemia. Pancreatitis or malabsorption
HYPOPARATHYROIDISM, TYPE IB at disorders including inflammatory bowel disease,
20q13.3), in which the metabolic findings of celiac disease, or resection of parts of the GI tract
hypoparathyroidism occur, but the general phe- can also lead to hypomagnesemia.
notype is normal except for the skeleton, and no Magnesium deficiency may result from renal
mutation is found in the GNAS1 gene. In pseudo- disease (either with (#248250 ICD+
hypoparathyroidism type II, an autosomal reces- HYPOMAGNESEMIA 3, RENAL; HOMG3) or
sive condition (%203330 ICD+PSEUDOHYPO without other tubular defects) or intestinal mal-
PARATHYROIDISM, TYPE II; PHP2), eleva- absorption which may occur in an autosomal
tion of urinary cAMP response in parathyroid recessive (#602014 ICD+HYPOMAGNESEMIA
hormone occurs but is diminished, but renal 1, INTESTINAL; HOMG1) as well as X-linked
phosphate excretion does not respond to PTH and patterns. Bartter syndrome refers to several auto-
hyperphosphatemia and hypocalcemia results. somal recessive disorders characterized by
Progressive osseous hyperplasia (#166350 decreased sodium chloride reabsorption in the
ICD+OSSEOUS HETEROPLASIA, PROGRE- thick ascending loop of Henle which leads to salt
SSIVE; POH) is an imprinted defect also due to a wasting; hypokalemic, hypochloremic metabolic
mutation resulting in loss of function of the alkalosis; and hypercalciuria with increased
Gs-alpha isoform of the GNAS gene (139320) on plasma renin activity and hyperactivity of the
the paternal allele. This condition presents with renin–angiotensin system with increased plasma
heterotopic bone formation in the dermis (oste- aldosterone; hypomagnesuria is often found as
oma cutis) and subcutaneous fat along with joint well. There are antenatal severe forms of Bartter
ankyloses and growth retardation of the limbs syndrome (#601678 ICD+BARTTER
affected by heterotopic bone formation. It is SYNDROME, ANTENATAL, TYPE 1) and
hypothesized that POH is an example of the (#241200 ICD+ BARTTER SYNDROME,
extreme presentation of PPHP. ANTENATAL, TYPE 2) as well as forms
The therapy for pseudohypoparathyroidism (#602522 BARTTER SYNDROME, TYPE 4A)
is the same as that for hypoparathyroidism and (#613090 BARTTER SYNDROME,
described earlier. TYPE 4B) associated with sensorineural deaf-
138 7 Disorders of Calcium Metabolism and Bone

ness. Replacement oral magnesium oxide given Activating Mutations of the Calcium-
several times daily will reverse hypoparathy- Sensing Receptor
roidism, hypocalcemia, and tetany, as hypo- Activating mutations of CaSR factitiously signal a
magnesemia resolves. Gitelman’s syndrome state of excessive calcium in the absence of excess
(#263800 ICD+ GITELMAN SYNDROME) is calcium which leads to a decrease in parathyroid
an autosomal recessive disorder associated hormone secretion and results in mild to moderate
with renal potassium and magnesium wasting hypocalcemia. (#601198 HYPOCALCEMIA,
leading to polydipsia with hypomagnesemia, AUTOSOMAL DOMINANT 1; HYPOC1) This
hypocalciuria, and hypokalemic alkalosis. condition can lead to calcium deposition in the
Patients may demonstrate generalized muscle kidney and in the central nervous system.
weakness, muscle cramps, tetany, seizures, par- Antibodies directed to the calcium-sensing recep-
esthesias, and episodic paralysis after strenu- tor may produce this same clinical presentation
ous exercise. and may be found in autoimmune hypoparathy-
Treatment of hypomagnesemic states is roidism as an associated finding. Antibodies to the
accomplished by oral or intravenous magnesium calcium-sensing receptor may also produce
administration. Intravenous or intramuscular hypercalcemia.
administration of a 50 % magnesium
(MgSO4.7H2O) sulfate at a dose of 0.05–0.2 mL/ Hyperphosphatemia
kg which provides 2.5–5 mg/kg elemental mag- Hyperphosphatemia may occur because of con-
nesium can be administered in emergency situa- genital defects or acquired conditions. Elevated
tions including convulsions as might be found in phosphorus will tend to complex with calcium
a neonate as long as careful cardiac monitoring leading to hypocalcemia and ectopic calcium
accompanies infusion since the EKG changes of phosphate crystal deposition and ectopic tissue
acute hypermagnesemia include a prolonged PR mineralization of subcutaneous tissues or of the
interval, increased QRS duration, and increased juxta-articular muscle. With acute phosphorous
QT interval and may lead to complete heart block administration either oral, intravenous, or rectal in
and cardiac arrest. Oral therapy may be accom- the form of enemas, hypocalcemia may be severe
plished with 50 % MgSO4.7H2O at a dose of enough to cause positive physical findings.
0.2 mL/kg/day. Familial hypophosphatemic tumoral calcinosis
Elevated serum magnesium may be asymp- (#211900 TUMORAL CALCINOSIS,
tomatic or may be associated with central ner- HYPERPHOSPHATEMIC, FAMILIAL; HFTC)
vous system depression and hypotonia. While is an autosomal recessive condition due to
decreased serum magnesium leads to impaired mutations in the UDP-N-acetyl-alpha-D-
parathyroid hormone secretion so does exces- galactosamine:polypeptide N-acetylgalactos-
sively elevated magnesium levels. A neonate of a aminyltransferase 3 gene (GALNT3,
mother who has been treated with significant 601756.0001) (GALNT3), FGF23 gene, or the
magnesium will itself have hypermagnesemia klotho gene (KL). Hyperphosphatemia occurs
and may also suffer from hypocalcemia. Other because of increased tubular resorption of phos-
sources of excessive magnesium include phate. Variation in findings may occur due to the
administration of IV or oral magnesium products different genes involved, but characteristically
including antacids or with enemas. there is a normal to elevated serum 1,25-dihy-
Hypermagnesemia is treated with increased droxycholecalciferol, normal serum calcium, and
hydration to excrete the magnesium load, but in normal serum PTH. FGF23 is characteristically
situations of hypocalcemia and hypoparathyroid- low while C-terminal variants of the full PTH mol-
ism, administration of calcium and ecule have been found to be high in some patients.
1,25-dihydroxyvitamin D may be necessarily on Nephrocalcinosis is likely along with precipitation
a temporary basis. of calcium phosphate crystals in subcutaneous
Hypocalcemia 139

tissue; periarticular calcification; pain in joints noted on the flow chart (Fig. 7.2), the level of
including the hip, elbow, and shoulder due to ecto- phosphorus gives much information, as if it is
pic calcification; and pain and swelling of long high, a deficiency of PTH or an inability to
bones, occur among other findings. respond to PTH is likely and if low, a deficiency
Hypocalcemia may occur during treatment for of vitamin D or its action is likely. The differen-
neoplasms due to tumor cell lysis. Tumor lysis tial diagnosis of various forms of pseudohypo-
syndrome (TLS) includes hyperphosphatemia, parathyroidism may require the measurement of
hyperkalemia, hypocalcemia, and hyperuricemia. plasma and urinary nephrogenic cAMP. The
Cell destruction from hemolytic anemia or crush diagnostic process may be complex, so refer to
injuries among other causes will lead to a similar Figs. 7.1 and 7.2 as well as to the descriptions of
presentation as tumor lysis syndrome. the various conditions.
Rhabdomyolysis can also lead to hyperphosphate-
mia and hyperkalemia; there may be either hyper-
or hypocalcemia encountered in rhabdomyolysis. Normal Bone Growth
Hyperphosphatemia found in chronic kidney
disease may require treatment by phosphate Bone is a complex organ which is affected by
binding agents. Older agents are calcium- or various endocrine factors but also produces its
aluminum-containing medications, while newer own endocrine agents. Bone is not a static organ
agents are non-calcium- or aluminum-based but once formed undergoes remodeling through-
agents (sevelamer hydrochloride compounds and out life. Bone is composed of an organic matrix,
lanthanum carbonate); they are more expensive, osteoid, comprised of protein which is mainly col-
and while they have theoretical benefits in con- lagen which becomes mineralized with the depo-
trolled studies, the data do not yet support their sition of calcium, phosphate, and hydroxyapatite.
preferential use. Bone mineralization begins with the formation
Ectopic bone formation is different than soft of the anlagen which is a framework of cartilage
tissue calcification described above. Ectopic bone upon which the future skeleton will be formed by
formation may occur due to hereditary conditions a process known as endochondrial ossification.
or after surgery or injury. Ectopic bone formation Chondrocytes progress through sequential stages
or heterotopic ossification is the formation of of resting, proliferating, and pre-hypertrophic
endochondral bone in soft tissues including mus- and hypertrophic growth leading to enlargement
cle, subcutaneous tissue, and fibrous tissue adja- of the anlagen. Hypertrophic chondrocyte cells
cent to joints. Hereditary progressive osseous toward the center of the cartilage produce colla-
hyperplasia (#166350 ICD+ OSSEOUS gen X and other factors in the mature extracellular
HETEROPLASIA, PROGRESSIVE; POH) is an matrix. Hypertrophic chondrocytes also produce
autosomal dominant condition caused by loss-of- angiogenesis factors, vascular endothelial growth
function mutations in the GNAS gene on the factor (VEGF), which promote the development
paternal allele which are the same gene defects of blood vessels in the area which subsequently
found in pseudopseudohypoparathyroidism. allow osteoblasts (bone-forming cells) and the
POH may be a condition of the extreme end inactive form osteocytes, osteoclasts (resorbing
of the range of manifestations of pseudo- cells), and hemopoietic cells to establish in the
pseudohypoparathyroidism. area. This forms a primary ossification center. The
hypertrophic chondrocytes undergo apoptosis and
are replaced by osteoblasts, formed out of mesen-
Diagnosis of Hypocalcemia chyme, which will add their secretions to the
extracellular matrix and will form trabecular bone.
The diagnosis of the etiology of hypocalcemia is Trabecular bone is porous and is found in the end
accomplished by the measurement of various of long bones, in vertebrae, and in flat bones such
minerals, hormones, and often vitamin D. As as the pelvis. In contrast cortical bone is dense and
140 7 Disorders of Calcium Metabolism and Bone

is found in the shaft of long bones and in the outer Osteoblasts as well as other cells produce
shell around cancellous bone at the end of joints receptor activator of nuclear factor kappa-B
and the vertebrae. Periosteum covers the outside ligand (RANKL) which is a member of the tumor
surface of long bones except at joints, while end- necrosis family of molecules. RANKL is located
osteum coats the inside of long bones. Bone on the surface of osteoblasts and interacts with the
growth in width occurs at the periosteum. osteoclastic receptor activator of nuclear factor
The extracellular matrix contains a host of kappa-B (RANK) to stimulate osteoclastic bone
collagen molecules including collagen type I n resorption. Osteoprotegerin (OPG) is also a
which contains α1(I) and α2(I) chains which are member of the tumor necrosis factor family and
coded by the genes COL1A1 and COL1A2; is a decoy receptor for RANKL and so inhibits
mutations in these genes lead to osteogenesis activation of osteoclasts. OPG production is
imperfecta. Mutations of other types of collagen stimulated by estrogen and by the medication
in the matrix lead to disorders that range in effect strontium ranelate. Denosumab is a medication
from lethality to dwarfism to early-onset arthritis. with a structure similar to OPG which is used to
A peripheral tube forms around the central carti- decrease bone resorption.
lage known as the diaphysis area. The primary On the other hand glucocorticoids increase
ossification center resides within this tube. Bone bone resorption by stimulating osteoclastogene-
marrow is formed centrally with the arrival of the sis and decrease osteoblastic function leading to
hemopoietic cells. At the ends of the cartilage, a net loss of bone and decrease in bone forma-
known as the epiphyses, secondary ossification tion. Bone resorption is also stimulated by
centers develop, and the growth plate, which will interleukin(s)-1, -6, and -11 (IL-1, IL-6, IL-11)
later appear as a lucency on radiograph of the and tumor necrosis factor (TNF) alpha and beta
ends of the bone and is responsible for longitudi- by enhancing osteoclast development; these
nal bone growth, is established in the epiphysis. agents also inhibit bone formation.
The epiphyses ultimately fuse at the end of nor- Bone resorption is inhibited with by estrogens
mal growth, a process stimulated by estrogen. and oral selective estrogen receptor modulators.
The metaphysis is the area of widening between Androgens also inhibit bone resorption, and while
the narrow diaphysis and the wider epiphysis. direct effects have been demonstrated, most of the
Further processes of chondrocyte proliferation, effect appears to be through the aromatization of
hypertrophy, and apoptosis progress allowing testosterone to estrogen. Calcitonin will also
remodeling and the deposition of calcium, phos- inhibit bone resorption and appears not to have a
phate, and hydroxyapatite (Ca10(PO4)6(OH)2) major physiologic function in this regard.
upon the extracellular matrix which provides
strength and stiffness of bone. The collagen in
bone provides elasticity and decreases the risk Vitamin D and Calcium Deficiency
of fracture. Thus, bone demonstrates strength
and a degree of elasticity. Disorders of bone may Hypocalcemia due to calcium deficiency, with or
arise from defects in calcification or defects in without rickets, had become uncommon in our era,
protein production. but more recently a reappearance of vitamin
Bone formation in remodeling occurs because D-deficient rickets in breast-fed infants not supple-
of physical and endocrine factors. Mechanical mented with vitamin D occurred. This can also
loading stimulates skeletal ossification and regu- occur in an infant receiving less than 33.8 oz or
lates changes in bone geometry and density. It is about 1 quart of vitamin D-supplemented infant
important that children are active for this reason. formula every day. Hypocalcemia of vitamin D
Bone formation is enhanced with growth hormone deficiency can present to such a severe degree that
and the IGF axis as well as thyroid hormone. hypocalcemic seizures occur. An infant raised on
While continuous exposure to parathyroid hor- an inappropriate fad diet deficient in vitamin D or
mone increases bone loss, intermittent exposure calcium or with the substitution of nondairy
stimulates bone formation. creamer for a milk-based formula, a situation that
Hypocalcemia 141

may arise in families with low income and may be frontal bossing, and more rarely craniotabes
considered a form of urban kwashiorkor, may (ping-pong ball-like skull which should not be
develop hypocalcemia. In cases in which calcium demonstrated clinically by the examiner) are
deficiency is the etiology and not vitamin D defi- found. Appetite is poor, walking is delayed, and
ciency, serum 25-hydroxyvitamin D is normal, but growth is decreased in many affected children.
1,25(OH)2D is either normal or high and PTH val- Radiographic findings of thin bones with cupping
ues are high. (concave), widening, and irregular “moth-eaten”
appearance of the metaphyses are diagnostic after
the condition is established. Costochondral swell-
Rickets (Table 7.2) ing is noted. Bone density may be decreased. In
those patients with hypocalcemia, all the findings
Rickets is a disorder of mineralization compared listed earlier for low calcium may be found.
to osteoporosis which is a disorder of protein for-
mation. Inadequate vitamin D intake or absorp- Nutritional Rickets
tion during childhood will cause inadequate Milk and other food products have been fortified
absorption of calcium and phosphorus, leading to with ergocalciferol in the USA, whereas other
hypocalcemia and hypophosphatemia (Table 7.2). areas of the developed world may provide vita-
These factors cause increased PTH secretion, min D to children as a capsule, so vitamin
which leads to increased bone turnover and ele- D-deficiency rickets is rare today. However, not
vated serum alkaline phosphatase activity. all dairy products are supplemented with vitamin
Chondrocytes in the growth plate of long bones D, and some children are given milk substitutes
and osteoid, the collagen-containing organic that have no vitamin D; inspection of the package
matrix of the bone trabeculum, fail to mineralize is necessary to determine the vitamin D content.
normally in this situation, causing rickets. Both Further, fad or well-intentioned but incorrectly
rickets and osteomalacia are characterized by constructed vegetarian/vegan diets may be defi-
decreased mineralization of osteoid; the former cient in vitamin D and require supplementation.
occurs during periods of growth when there are Alternatively, as normal breast milk has less
open epiphyses, causing abnormal formation of vitamin D content than recommended for an
long bones, ribs, and skull, and the latter occurs infant, especially if the mother herself is vitamin
after growth ceases in the adult and leads to a ten- D deficient, an infant raised on breast milk with-
dency toward fractures. Radiologic evaluation of out vitamin D supplementation may be deficient.
the bones shows undermineralization of the This problem is intensified with the use of sun-
growth plate, an indistinct and ragged metaphysis, screen, an important measure to decrease the risk
and bowing due to the influence of weight bearing of skin cancer, and is further enhanced in a child
on bones lacking tensile strength. Rickets is often with a dark complexion. There is a resurgence of
divided into calciopenic and phosphopenic rickets rickets in infancy due to these nutritional issues
based upon which ion deficit is primary. even in a sunny climate. If an older child is not
Findings in nutritional rickets are often exposed to the sun (e.g., because of institutional-
encountered when the child enters the toddler ization or complete swaddling covering the skin),
stage and include genu valgum (“knock knees”) the conversion of 7-dehydrocholesterol to vita-
or genu varum (“bow leg”) developing after the min D3 will be limited, and vitamin D deficiency
child begins walking and usually associated with results. Children with dark skin color living at
pain in ambulation. Bowing of the arms, classic northern latitudes, wearing opaque clothes, and
and often palpable widening of the metaphyses of mostly remaining indoors are at particular risk
the long bones, “rachitic rosary” which appears as for developing rickets if not supplemented with
an inverted “V” parallel and lateral to the costal vitamin D.
margins, Harrison grooves or sulci (indentation Fat malabsorption due to a variety of condi-
of the lower chest wall as a result of softening), tions may lead to vitamin D deficiency and
142

Table 7.2 Types of rickets


Serum Serum Serum Serum
Defect Serum Ca PO4 alk phos 25(OH)D 1,25(OH2)D Serum PTH Other features
Vitamin D deficiency Normal at first, then may Normal High to Low Low to Normal at Was rare in the developed world owing to
rise after parathyroid at first, extremely normal first, then vitamin D supplementation. Now resurging
hormone secretion then low high high due to breast-feeding without adequate
increases vitamin D supplementation. Occurs in fat
malabsorption, deficient maternal vitamin D,
prematurity, sunlight avoidance
Hepatocellular rickets Low Low High Low Low to High
normal
Anticonvulsant-associated Low Low High Low Low to High Due to increased metabolism of 25(OH)D
rickets normal (e.g., from phenobarbital and hydantoins in
addition to lack of sunlight exposure,
usually in a poorly ambulatory individual)
Vitamin D-dependency rickets Low Low or High Normal Low High Requires high doses of vitamin D. AR
7

type 1 or pseudovitamin D- normal inheritance due to a loss-of-function


deficiency rickets mutation of the 1α-hydroxylase gene
Vitamin D-resistant rickets or Low Low High Normal High High Might have alopecia, growth failure, dental
vitamin D-dependency rickets hypoplasia. AR inheritance due to
type 2 loss-of-function mutation of the vitamin D
receptor
X-linked hypophosphatemic Normal Low High Normal Low Nl to high X-linked inheritance due to mutation of
rickets PHEX gene
From: Fisher DA, ed. The Quest Diagnostics Manual: Pediatric endocrinology. San Juan Capistrano: Quest Diagnostics Incorporated 2000
AR autosomal recessive, PTH parathyroid hormone, Nl normal
Disorders of Calcium Metabolism and Bone
Hypocalcemia 143

rickets. This may occur with disturbance of the vitamin D metabolism: (a) fat malabsorption due
enterohepatic circulation and absence of pancre- to inadequate bile salt in the small intestine
atic enzymes or may follow bariatric surgery. decreases absorption of the fat-soluble vitamin
Patients with cystic fibrosis or short bowel syn- D; (b) hepatic 25-hydroxylation may be
drome are subject to vitamin D malabsorption. A decreased; and (c) because vitamin D is excreted
calcium-deficient diet or, more rarely, inadequate in bile, the enterohepatic circulation of vitamin D
dietary phosphate also can lead to rickets. Diets may be interrupted. Therapy with 1,000–2,000 IU
laden with phytates, found in some cereals, also (25–50 μg) of vitamin D daily may cure this form
will lead to calcium deficiency because they bind of rickets, but the use of oral 25(OH)D may be
calcium in the GI tract and limit absorption. necessary to treat children with biliary atresia.
There are three phases of vitamin D-deficient Exposure to the sun may be beneficial in such
rickets (according to C. Arnaud): at first serum patients as well but this should not be excessive as
calcium is low (this phase may be missed at the noted elsewhere.
onset) which leads to elevation of PTH that raises
serum calcium by mobilization from the bone. Renal Osteodystrophy
The response to parathyroid hormone ultimately In significant renal disease, impaired 1-hydroxyl-
falters due to a form of PTH resistance, and ation of 25 OH vitamin D will lead to the hypo-
serum calcium falls again, while serum phos- calcemia that triggers PTH secretion causing
phate rises in the second stage. This is a time in secondary hyperparathyroidism. The PTH will
which hypocalcemic seizures may be manifest. increase remodeling of the bone, and serum alka-
The hypocalcemic seizures found in rickets may line phosphatase will rise. The serum phosphorus
be accompanied by hyperphosphatemia and ele- remains elevated because of decreased urinary
vated PTH falsely suggesting pseudohypopara- excretion caused by the renal disease. Thus, renal
thyroidism. Rising PTH levels finally overcome osteodystrophy results in osteitis fibrosa cystica
the resistance and cause hypophosphatemia with caused by secondary hyperparathyroidism, as
repletion of serum calcium with further resorp- well as renal rickets or osteomalacia due to bone
tion of bone in the third stage. Alkaline phospha- undermineralization. Bowing of the lower
tase rises, and the characteristic radiographic extremities, bone pain, and myopathy may occur.
features of rickets begin. If vitamin D is inade- Parathyroid hormone values are increased due to
quate, calcium and phosphorus both fall, while elevated serum phosphorus. Therapy involves
PTH and alkaline phosphatase rise further and 1,25-dihydroxyvitamin D, which increases serum
severe rickets develops. The changes of vitamin ionized Ca2+, which in turn decreases PTH
D-deficient rickets can be reversed with vitamin secretion. Oral calcium carbonate is given with
D treatment. meals to block intestinal phosphate absorption,
Gastric bypass surgery is now more frequently and a low-phosphate diet is recommended.
invoked in the treatment of severe obesity in Phosphate binding agents may be necessary in
adolescence. The side effects of gastric bypass secondary hyperparathyroidism of renal disease
surgery can be vitamin D deficiency and meta- and are mentioned below in hyperphosphatemia.
bolic bone disease. This may be due to malab- Because renal disease may be subtle in some
sorption, dumping syndrome, or absence of cases, evaluation of renal function is important in
pancreatic enzyme or interruption of the entero- the evaluation of hypocalcemia.
pathic circulation depending upon the surgical
procedure invoked. Anticonvulsant-Associated Rickets
or Hypocalcemia
Hepatocellular Rickets Anticonvulsants such as phenobarbital and
Liver diseases such as biliary atresia, neonatal hydantoins speed the metabolism of 25(OH)D to
hepatitis, some hereditary liver diseases, and, more inactive metabolites. Because some
rarely, cystic fibrosis cause several difficulties with children taking these substances may be
144 7 Disorders of Calcium Metabolism and Bone

institutionalized, a decreased intake of dairy rickets, leads to the inability to produce


products fortified with vitamin D also may occur, 1,25(OH)2D by 1-hydroxylation. This condition
as well as reduced sunlight exposure. These com- may be noted clinically at age 12–16 weeks and
bined effects may lead to rickets or hypocalce- is inherited in an autosomal recessive pattern.
mia. This complication is unusual in ambulatory Besides nondetectable or decreased serum cal-
children receiving anticonvulsants. citriol, there will also be hypophosphatemia,
Treatment with 800 IU (20 μg) of vitamin D increased serum PTH, increased serum alkaline
daily is appropriate to correct hypocalcemia and phosphatase, and generalized aminoaciduria.
hypophosphatemia in these children, especially if Patients with this condition require very large
adequate calcium intake is assured. doses of vitamin D (200,000–1,000,000 IU
daily) or physiologic doses (0.5–1.0 μg) of
Rickets of Prematurity 1,25(OH)2D to cure the rickets. The levels of
Very low birth weight preterm infants have not 25(OH)D are normal or high and of 1,25(OH)2D
received the normal third trimester placental are low; the latter do not increase during treatment
transfer of calcium (150 mg/kg body weight/day) with vitamin D.
or phosphate (75 mg/kg/day), and after birth, Vitamin D-dependency rickets, type 2 (#277440
their intestine may not be adequate to absorb cal- VITAMIN D-DEPENDENT RICKETS, TYPE
cium to replace the interrupted placental func- 2A; VDDR2A at 12q12-q14O) or vitamin D-
tion. Further, diminished 25-hydroxylation resistant rickets is inherited in an autosomal reces-
occurs because of liver immaturity, even if the sive pattern and is due to inactivating mutation of
mother was supplemented with vitamin D and the the vitamin D receptor, leading to a defect in the
infant is not deficient in vitamin D. Hypocalcemia, binding of 1,25(OH)2D to its nuclear receptor.
hypophosphatemia, elevated alkaline phospha- These patients have an extremely elevated serum
tase activity, rib and limb fractures, and rachitic concentration of 1,25(OH)2D (exceeding 150 pg/
changes at the diaphyses and metaphyses of long mL; normal, 15–90 pg/mL depending upon age),
bones will result. Serum concentrations of but endogenous 1,25(OH)2D and normal doses
1,25(OH)2D are extremely high as a compensa- of exogenous 1,25(OH)2D are ineffective.
tory mechanism, exceeding 120 pg/mL (normal Treatment often requires remarkably high doses of
values are 15–90 pg/mL depending upon age); 1,25(OH)2D, in excess of 10 μg/day.
these elevated values of 1,25-dihydroxyvitamin Additional features include cyst of the skin
D do not indicate that the defect is resolving and alopecia sparing the eyebrows. Patients have
however as 25-hydroxy-vitamin D levels indi- hypotonia and difficultly in walking as well as
cate vitamin D stores, while 1,25-dihydroxyvi- delayed motor development, and seizures may
tamin D levels do not. If the infants also have occur due to hypocalcemia. There is secondary
bronchopulmonary dysplasia and are receiving hyperparathyroidism which leads to hypophos-
long-term loop diuretic therapy, they will lose phatemia and increased serum alkaline phospha-
more calcium through hypercalciuria. tase levels, but since vitamin D receptors are
Treatment consists of the use of calcium and inactive, there is hypocalcemia. Patients also may
phosphate supplements in the infant’s milk, the be found with delayed tooth eruption and enamel
use of high-mineral formulas, and additional hypoplasia with early onset of dental caries.
vitamin D administration. Rickets, hypocalcemia, enamel hypoplasia,
and signs of secondary hyperparathyroidism are
Genetic Causes of Rickets seen in these children as are short stature and
total alopecia.
Defects in Vitamin D Production or Action
1α-hydroxylation deficiency rickets (*264700 Hypophosphatemic Rickets
PSEUDOVITAMIN D DEFICIENCY RICKETS Hypophosphatemia characteristically is due to
at 12q14), also called vitamin D-dependency renal phosphate wasting. There are often no
rickets type 1 and pseudovitamin D-deficiency symptoms of hypophosphatemia but occasionally
Hypocalcemia 145

patients complain of weakness, bone pain, rhab- adult height. Clinical studies have just begun on
domyolysis, and altered mental state. However, FGF23 with as a treatment for X-linked hypo-
the secondary effects of hypophosphatemia phosphatemic rickets.
involve disorders of bone mineralization. The There are autosomal recessive forms of hypo-
most common form of inherited rickets in the phosphatemic rickets due to other gene defects
developed world is X-linked hypophosphatemic (#241520 ICD+ HYPOPHOSPHATEMIC
rickets (*307800 HYPOPHOSPHATEMIA, RICKETS, AUTOSOMAL RECESSIVE, 1;
X-LINKED at Xp22.2-p22.1), an X-linked domi- ARHR1 due to a homozygous mutation in the
nant condition found in hemizygous males and DMP1 gene at 4q21.) which may include nerve
heterozygous females. Affected children have a deafness in the constellation of symptoms and
reduction in the amount of phosphate reabsorbed autosomal recessive forms without nerve deaf-
in the nephron and lose considerable phosphate in ness (#613312 HYPOPHOSPHATEMIC
their urine. A reduction in the serum value of RICKETS, AUTOSOMAL RECESSIVE, 2;
1,25(OH)2D also occurs. Affected children have ARHR2- due to a homozygous mutation in the
hypophosphatemia, causing insufficient mineral- ENPP1 gene at 6q 23.2) and autosomal dominant
ization of the bone, even though their serum cal- forms (#193100 ICD+ HYPOPHOSPHATEMIC
cium is normal. By the time the child begins to RICKETS, AUTOSOMAL DOMINANT;
walk, bowing of the lower extremities is seen, but ADHR due to a mutation in the FGF23 gene at
unlike those with other forms of rickets, affected 12p13) of hypophosphatemic rickets in addition
children do not have tetany, myopathy, a rachitic to the more frequent X-linked dominant form.
rosary, or secondary hyperparathyroidism. Treatment of hypophosphatemic rickets aims
These children have inadequate dental enamel to promote bone healing clinically and on radio-
and dental cysts leading to frequent tooth decay. graphs and reduction of alkaline phosphatase to
Other physical features include midfacial hypo- normal levels. Phosphate is administered in mul-
plasia and frontal bossing. The disorder is usu- tiple daily doses since values drop quickly after
ally due to a mutation in the PHEX gene on ingestion. Sixty mg/kg/day is the usual dose for
Xp22.1.p leading to elevations of FGF23. younger children and 40 mg/kg/day for adoles-
Treatment involves oral phosphate supplements cents. Gastrointestinal distress or diarrhea may
in up to five divided doses to attempt to maintain result. Measuring urinary phosphorus is a way of
serum phosphate values within normal limits checking compliance as it will rise with treat-
most of the day, although oral phosphate can ment even further than the baseline state in this
precipitate diarrhea. Children with X-linked condition but will not rise if the child is not tak-
hypophosphatemic rickets have a decreased ing phosphate. Due to impairment of 1 alpha
ability to produce 1,25-dihydroxyvitamin D2 to hydroxylase, 1,25-dihydroxyvitamin D is admin-
an incomplete defect in the regulation of istered in doses of 0.5–1 μg/day until the pubertal
25-hydroxyvitamin D-1-alpha-hydroxylase growth spurt when a dose of 0.5–1.5 μg/day
activity. In states of hypophosphatemia might be necessary. Monitoring urinary calcium
1,25-dihydroxyvitamin D should increase but is is necessary to avoid the development of hyper-
inappropriately normal in X-linked hypophos- calciuria which may be accompanied by nephro-
phatemic rickets. Increased levels of parathy- calcinosis. A urinary calcium/creatinine ratio
roid hormone do not overcome this block, but should be less than 0.35 mg/mg creatinine in
there is a normal response to calcitonin admin- toddlers and less than 5 mg/kg of body weight
istration in clinical studies, but calcitonin is not per day in older children. Ultrasonography to
currently used for therapy. 1,25(OH)2D is admin- determine if there is nephrocalcinosis is carried
istered in doses of 0.25–0.75 μg every 12 h, as out every year or 2. It is also recommended to
the patients have inadequate amounts of serum supplement with ergocalciferol or cholecalciferol
1,25(OH)2D for the degree of hypophosphate- to raise 25-hydroxy-vitamin D levels to normal.
mia. Some clinical studies used GH treatment in A rare paraneoplastic condition which
affected subjects, but there was no effect on presents with biochemical findings similar to
146 7 Disorders of Calcium Metabolism and Bone

hypophosphatemic rickets is oncogenic osteoma- are the urinary fractional excretion, tubular
lacia. This condition is usually associated with a resorption, and tubular threshold for phosphate.
phosphaturic mesenchymal tumor, mixed con-
nective tissue variant, which can rarely occur at Treatment of Vitamin D Deficiency
any age and gender and often is undiscovered for (Table 7.3)
years due to the small size of the tumor. FGF23 is There is controversy but for children a serum
elevated, and phosphaturia occurs and ultimately 25(OH)D value is considered normal if D ≥20 ng/
rickets. mL (50 nmol/L), while 15–<20 ng/mL (37.5 and
The bone cyst found in McCune–Albright 50 nmol/L) is vitamin D insufficiency and
syndrome may lead to elevated FGF23. This can ≤15 ng/mL (37.5 nmol/L) is vitamin D defi-
lead to hypophosphatemic rickets or osteomala- ciency; a reasonable goal in children is attain-
cia. Thus, phosphate levels must be evaluated in ment of 20–30 ng/mL, but these values may be
patients with McCune–Albright syndrome espe- revised as research continues into the optimal
cially before treatment of bone cysts. dose. Obese children tend to have a lower value
Bisphosphonates are invoked as experimental of vitamin D, but it is not clear what are the clini-
treatment for McCune–Albright syndrome but cal implications of these lower levels or whether
are not proven to be beneficial as yet; if used dur- the vitamin D is sequestered in fat. The Institute of
ing hypophosphatemic conditions, phosphate Medicine of the USA recommends 400 IUs of
would be expected to drop further causing vitamin D to be administered to infants up to 1
increased complication. year of age and due to a high prevalence of sub-
clinical vitamin D deficiency which is heralded
Renal Tubular Acidosis by increased parathyroid hormone values in the
Proximal renal tubular acidosis is due to inability USA has increased recommendations to 600 IU/
to resorb bicarbonate in the renal tubule and is day between the ages 1 and 18 years. These dos-
often associated with the Fanconi syndrome (in ages are used for maintenance rather than treat-
which glucose, amino acids, uric acid, and phos- ment of deficiency. Treatment of vitamin
phate are also not reabsorbed). Elevated parathy- D-deficient children with ergocalciferol at
roid hormone may lead to secondary Fanconi 3,000 U daily for infants less than 6 months and
syndrome with a mild acidosis in vitamin 6,000 U daily for age 6 months to 12 years for 6
D-deficient rickets before treatment with vitamin weeks is usually curative, although additional
D; this situation may falsely suggest primary renal calcium intake may be required. In cases of mal-
tubular acidosis. Distal renal tubular acidosis is absorption due to gastrointestinal disease (cystic
associated with an inability to acidify urine. These fibrosis, short bowel syndrome, post-pediatric
patients present a manner similar to vitamin surgery), increased doses of vitamin D are needed
D-deficient rickets except that in this case there is with careful monitoring to avoid overtreatment.
hypercalciuria which may lead to nephrocalcino- Excessive vitamin D intake leads to hypervita-
sis and renal failure. Treatment with vitamin D minosis D, and hypercalcemia may develop, and
will worsen the situation by increasing hypercalci- since vitamin D is fat soluble, it will take weeks
uria, but treatment with bicarbonate will help or months (6 months or even more) to return to
remedy this situation. normal stores; thus monitoring of 25-hydroxy-
vitamin D levels is important during treat-
Diagnosis of Rickets ment. There are reports of hypervitaminosis D in
The differential diagnosis of rickets is made by children treated within the recommended guide-
measuring serum Ca, PO4, and increased alkaline lines; thus every child must be individualized by
phosphatase determinations, as well as serum monitoring vitamin D, alkaline phosphatase, and
25-hydroxyvitamin D. Parathyroid hormone is PTH values to determine whether the deficiency
elevated in many of the stages of rickets. Albumin is repleted. To avoid overtreatment, monitoring
and creatinine are also useful measurements as of serum calcium and urinary calcium which rises
Hypercalcemia 147

Table 7.3 Calcium and vitamin D dietary reference intakes by life stage
Calcium Vitamin D
RDA (mg/day) RDA (IU/day) Serum 25OHD
(intake that covers (intake that covers level (ng/mL)
Life-stage group (age needs of ≥97.5 % of UL needs of ≥97.5 % (corresponding
and gender) population) (mg/day)a of population) to the RDA)b UL (IU/day)a
1–3 years (M + F) 700 2,500 600 20 2,500
4–8 years (M + F) 1,000 2,500 600 20 3,000
9–13 years (M + F) 1,300 3,000 600 20 4,000
14–18 years (M + F) 1,300 3,000 600 20 4,000
19–30 years (M + F) 1,000 2,500 600 20 4,000
31–50 years (M + F) 1,000 2,500 600 20 4,000
51–70 years (M) 1,000 2,000 600 20 4,000
51–70 years (F) 1,200 2,000 600 20 4,000
71+ years (M + F) 1,200 2,000 800 20 4,000
Pregnant or lactating (F)
14–18 years 1,300 3,000 600 20 4,000
19–50 years 1,000 2,500 600 20 4,000
Infants
0–6 months (M + F) 200c 1,000 400c 20 1,000
6–12 months (M + F) 260c 1,500 400c 20 1,500
Data from Dietary reference intakes for calcium and vitamin D,National Academies Press, 2010. Used with
permission
M male, F female
EARs for calcium were 500 mg/day for ages 1–3 (M + F), 800 mg/day for ages 4–8 and 19–50 (M + F) and ages 51–70
(M), 1,000 mg/day for ages 51–70 (F) and 71+ (M + F), and 1,100 mg/day for ages 9–18 (M + F). EAR for vitamin D
was 400 IU/day for all life-stage groups
a
UL indicates level above which there is risk of adverse events. The UL is not intended as a target intake (no consistent
evidence of greater benefit at intake levels above the RDA)
b
Measures of serum 25OHD levels corresponding to the RDA and covering the requirements of at least 97.5 % of the
population
c
Reflects AI reference value rather than RDA. RDAs have not been established for infants

in hypervitaminosis D is helpful. Use of must be carried out to make sure overtreatment


1,25-dihydroxyvitamin D is not recommended in does not occur.
states of vitamin D deficiency. One reason is that In some cases the rickets is so severe that
it would do nothing to replete the stores of the orthopedic surgery is required to correct the
body’s vitamin D. Secondly, if vitamin D treat- abnormalities.
ment is offered in vitamin D-deficiency rickets, Dihydrotachysterol is a vitamin D molecule
serum 1,25-dihydroxyvitamin D levels rise to that requires metabolism by the liver but not the
supraphysiologic levels which would not occur kidney. Thus, it may be preferred in situations of
with the replacement of 1,25-dihydroxyvitamin renal disease, and its rapid onset of action is also
D in standard doses itself. beneficial.
If compliance appears likely to be a problem,
“stoss” therapy may be invoked; this is one large
single dose of 150,000–600,000 units of vitamin D Hypercalcemia
given every 3 months as an oral dose divided
throughout 1 day or single intramuscular injection Hypercalcemic symptoms usually occur when
(IM injection is not available at the time of this calcium levels are greater than 12 mg/dL, but
writing in the USA however). Again monitoring the definition of hypercalcemia is a calcium
148 7 Disorders of Calcium Metabolism and Bone

Table 7.4 Causes of hypercalcemia


Serum Serum Alk 25(OH) 1,25(OH)
Condition Ca PO4 phos PTH vitamin D vitamin D Other features
Primary High Low High High Normal Nl or high
hyperparathyroidism for Ca
Familial High Normal, Normal Normal Normal Normal Urine Ca is low. Usually
hypocalciuric high, or or high or high benign but in homozygosity,
hypercalcemia low profound hypercalcemia
and diffuse
hyperparathyroidism
develops as above. AD
inheritance
Hypercalcemia of High Nl or High Low Normal Nl Due to secretion of PTHrP,
malignancy low which creates a state of
hyperparathyroidism
Hypervitaminosis D High Nl or Nl or Low High Nl or high Due to overtreatment or fad
low low megavitamin diets
Renal insufficiency Low Nl or Nl or High Normal Decreased Could lead to tertiary
and secondary high high autonomous (or tertiary)
hyperparathyroidism hyperparathyroidism
Increased High Normal Normal Normal Normal High Occurs in granulomatous
1α-hydroxylation of disease and neoplasms
25 OH vitamin D (lymphomas)
Immobilization High Normal High Nl Nl Nl
hypercalcemia to low
Hyperthyroidism High Nl/high Low Nl Nl or low
Adrenal High Nl Low Nl Nl or low
insufficiency
Thiazide diuretics High Nl Low Nl Nl or low Urine Ca is low
Hypervitaminosis A High Nl Low Nl Nl or low
Modified from: Fisher DA, ed. The Quest Diagnostics Manual: Pediatric endocrinology. San Juan Capistrano: Quest
Diagnostics Incorporated 2000 with permission
Nl normal, PTHrP parathyroid hormone-related peptide

level above the age-related normal range for the (nonpurulent conjunctivitis), shortening of the
laboratory which usually means above 10.8– QT/QTc due to shortening of the ST interval on
11.3 mg/dL (Table 7.4). Lethargy, weakness, EKG recording, and extremely wide QRS, low R
inability to concentrate, and depression may wave, disappearance of p waves and tall peaking
develop. Many patients have nausea, vomiting, T waves. In addition, hypertension, hypercalci-
anorexia, constipation, and weight loss; the neo- uria and nephrolithiasis, pancreatitis, and peptic
nate may manifest gastroesophageal reflux, leth- ulcer disease. Hypercalcemia also may lead to
argy, decreased weight gain, and lack of growth hypertension. If serum calcium increases to more
in length. Because a high extracellular calcium than 16 mg/dL, stupor and coma may occur.
concentration impairs the capacity of the distal Many nutritional and maternal causes of
tubule of the nephron to respond to ADH, hyper- hypercalcemia are seen in the newborn. Thus,
calcemic patients have polyuria and an inability excessive administration of calcium or, con-
to concentrate the urine; dehydration and azote- versely, inadequate administration of phosphorus,
mia may result as well as nephrolithiasis, neph- as well as excessive vitamin D administration
rocalcinosis, and calciuria. Signs on physical directly to the child or, rarely, through the moth-
examination may include band keratopathy of er’s breast milk, causes elevation of serum Ca.
the medial and lateral margins of the cornea Subcutaneous fat necrosis due to birth trauma or
Hypercalcemia 149

birth asphyxia elevates serum Ca due to the bone resorption, mainly of the distal phalanges;
1-hydroxylase activity of macrophages. osteitis fibrosa; fractures of the ribs and other
bones; peptic ulcer; and hypertension are found
in affected children. Because PTH stimulates the
Primary Hyperparathyroidism synthesis of 1,25(OH)2D by 1-hydroxylation,
serum 1,25(OH)2D is elevated but not
Hyperparathyroidism can lead to hypercalcemia, 25-hydroxyvitamin D as would occur in vitamin
but this condition is rarer in children than in the D toxicity. This diagnosis is made if serum ion-
adult. Hypercalciuria occurs with hyperparathy- ized calcium is elevated with inappropriately
roidism mainly due to the hypercalcemia and increased serum PTH, as PTH should be sup-
may lead to nephrolithiasis and even renal fail- pressed to nondetectable levels in a normal
ure. Bone pain, muscle aches, and arthropathy patient with elevated serum calcium.
are reported in pediatric series. In the postoperative state after parathyroidec-
Hyperparathyroidism may occur because of an tomy, the patient may experience hypocalcemia of
adenoma of one or more of the parathyroid glands the “hungry bone’s syndrome.” The resorption of
or because of mutations in the calcium-sensing the bones caused by elevated parathyroid hor-
receptor. Hyperparathyroidism might be an iso- mone abruptly ceases, and the bones then avidly
lated finding in a sporadic or autosomal dominant take up calcium lowering serum calcium levels.
pattern (#145000 HYPERPARATHYROIDISM 1; The calcium must be replaced by oral or intrave-
HRPT1) or can be found in the multiple endocrine nous method in the immediate period after such
neoplasia syndromes (MEN 1, MEN 2). However, surgery. The accompanying hypo-phosphatemia
since hyperparathyroidism may be the first mani- should however not be treated as administration of
festation of multiple endocrine neoplasia syn- phosphate will lower calcium further. Associated
dromes 1 and 2A, it is incumbent upon the with these changes are hypomagnesemia which
treating physician to eliminate the diagnosis of should be replaced and hyperkalemia which might
multiple endocrine neoplasia type I before also have to be addressed. As stated above, hungry
assuming the patient has isolated hyperparathy- bone syndrome is also found in replacement of
roidism (see Chap. 14). vitamin D after severe vitamin D deficiency.
Transient hyperparathyroidism in the neonate
may occur if the mother has hypoparathyroid-
ism. Maternal hypocalcemia causes increased Familial Hypocalciuric Hypercalcemia
fetal PTH secretion to maintain a normal fetal
serum calcium, but PTH secretion continues into Familial hypocalciuric hypercalcemia (FHH
the newborn period and causes elevated serum #145980 HYPOCALCIURIC HYPER-
Ca in the newborn for a few days to weeks. CALCEMIA, FAMILIAL, TYPE I; HHC1 at
Increased bone remodeling may be found since 3q13.3-q21) or (FHH) is an autosomal dominant
the condition was active during fetal period. condition due to inactivating mutation of the
Primary hyperparathyroidism causes increased CaSR which leads to a higher set point of the
absorption of ingested calcium and increased receptor in the parathyroid cells. Elevated
release of calcium from mobilized bone as well calcium values are seen by the sensor as normal,
as increased urinary excretion of phosphorus and and these inappropriately elevated values are
calcium. Thus hypercalcemia and hypophospha- maintained by increased PTH secretion, so that
temia occur, and, ultimately, nephrocalcinosis an equilibrium is reached at a higher than nor-
develops. Mild hypercalcemia leads to constipa- mal range of serum calcium (11–12 mg/dL is
tion and polyuria, whereas in those with more common) and ionized calcium. Parathyroid
may include significant hypercalcemia, manifes- hormone is either elevated or inappropriately
tations, arrhythmia, hypotonia to the level of normal for the degree of hypercalcemia.
respiratory compromise. Bone pain; subperiosteal Hypercalcemia, hypermagnesemia, hypocalciuria,
150 7 Disorders of Calcium Metabolism and Bone

and hypophosphatemia are found. Usually no Miscellaneous Causes


symptoms occur, but conditions due to the ele-
vated calcium including pancreatitis and choleli- Hypercalcemia of Malignancy
thiasis are found in some subjects. This condition Hypercalcemia may be caused by elevation of
may lead to inappropriate diagnosis of hyper- PTHrP in patients with malignancy. This may be
parathyroidism and an inappropriate removal of found in children with leukemia, lymphoma,
the parathyroid glands; the diagnosis may be rhabdomyosarcoma, Ewing sarcoma, and other
made in patients with a family pattern of elevated neoplasms. The treatment of the underlying
serum calcium in the face of decreased urinary malignancy usually reverses hypercalcemia.
calcium excretion. Affected patients will not
respond to a low-Ca diet, for they will mobilize Other Endocrine Disorders Causing
bone calcium to make up for the deficit and incur Hypercalcemia
more skeletal complications. Hyperthyroidism leads to active bone turnover
In contrast to heterozygous FHH which and enhanced resorption, causing mild hypercal-
requires no parathyroidectomy, homozygosity or cemia. Adrenal insufficiency allows increased
compound heterozygousity of the inactivating intestinal calcium absorption and may lead to
mutation of the CaSR leads to profound and hypercalcemia. Replacement with glucocorti-
potentially fatal hypercalcemia (14–20 mg/dL) in coids will reverse hypercalcemia in adrenal
a condition known as neonatal severe hyperpara- insufficiency.
thyroidism (#239200 HYPERPARAT
HYROIDISM, NEONATAL SEVERE; NSHPT) Drug-Induced Hypercalcemia
in which parathyroid hormone is extremely ele- Thiazide diuretics are natriuretic but not calci-
vated in spite of the severe hypercalcemia. uretic and cause diminished urinary calcium
Neonatal severe hyperparathyroidism does excretion and so may cause mild hypercalcemia.
require parathyroidectomy and may respond to They are used in conditions prone to developing
the use of bisphosphonates as calcium-lowering urinary calcium stones.
agents in preparation for surgery (see below). Hypervitaminosis D may occur if excess vita-
Urinary calcium is inappropriately low (less than mins are given to a child in the therapy for a hypo-
4 mg/kg/day), and the calcium/creatinine ratio is calcemic disorders or in a mistaken confidence in
usually less than 0.10 in spite of the level of megavitamin therapy. Fat-soluble vitamin D [ergo-
hypercalcemia. Severe bone resorption, frac- calciferol (vitamin D2) or cholecalciferol (vitamin
tures, and rachitic changes may be seen in those D3)] are stored for months in fat tissue, so an over-
who survive the neonatal period. dose is quite long lasting. However 1,25(OH)2D is
Hyperparathyroidism may be due to adeno- short acting, so that excessive dosage can be
mas or hyperplasia of the chief cells. quickly corrected. A discussion when to use either
Hyperparathyroidism might rarely be associated medication is noted above. Individual monitor-
with multiple ossifying fibromas of the maxilla ing must be carried out with the use of ergocalcif-
and mandible (#145001 HYPERPARATHY erol or cholecalciferol as vitamin D toxicity has
ROIDISM 2; HRPT2) which may also occur been reported in children who were treated with
with renal carcinoma or Wilms tumor, thyroid standard recommended doses. Glucocorticoids
carcinoma, and parathyroid carcinoma. (prednisone at 1–2 mg/kg/day) may be used to
Some infants with mild disease have remission reduce intestinal calcium absorption in the condi-
or a mild course, but the usual treatment for a tion. Children in England at the end of World War
parathyroid adenoma is surgical extirpation. II frequently manifested hypercalcemia, appar-
If hyperplasia of multiple glands is found, all of ently because of excess vitamin D intake.
the glands may be removed, with a fraction of Hypervitaminosis A increases bone resorption
one reimplanted in the forearm, where it can causing hypercalcemia and may occur with mega-
be surgically reduced in size if hypercalcemia vitamin therapy. Usually vitamin A excess is found
continues or recurs. with vitamin D excess if caused by fad diets.
Hypercalcemia 151

Milk Alkali Syndrome and Hypercalcemia Immobilization Hypercalcemia


of Renal Insufficiency A child (or more usually an adolescent) who is
Excessive ingestion of calcium usually in the immobilized, usually from a fracture and trac-
form of calcium carbonate is linked to this hyper- tion, will have enhanced bone resorption and
calcemic condition which can lead to renal fail- diminished bone formation, which may lead to
ure. Originally described with the use of calcium immobilization hypercalcemia. Intramuscular
and absorbable alkali for the treatment peptic calcitonin (50–200 U) will block bone resorption,
ulcers, it may now occur due to excess of calcium although ambulation is the best treatment.
intake for supplementation of dietary sources.
Individuals with renal insufficiency may Williams Syndrome
have hyperphosphatemia, hypocalcemia, Williams syndrome, now more frequently termed
decreased 1,25(OH)D production, and therefore Williams–Beuren syndrome (#194050
increased PTH secretion, causing secondary WILLIAMS-BEUREN SYNDROME; WBS at
hyperparathyroidism. This may become autono- 7q11.2), is a sporadic and contiguous gene dele-
mous (tertiary hyperparathyroidism). Thus, tion syndrome consisting of short stature and
even when calcium increases, it does not sup- developmental delay, supravalvular aortic steno-
press PTH secretion. Calcium carbonate, lac- sis, pulmonary arterial stenosis, and a pathogno-
tate, or bicarbonate intake given to block monic facial appearance; it is due to a mutation in
intestinal phosphate accumulation in chronic the gene for elastin. “Elfin facies,” consisting of a
renal insufficiency may lead to a form of the small head, protuberant ears, a cupid’s bow lip
milk alkali syndrome, which includes hypercal- with short philtrum, peg-like teeth, blue iris with
cemia, hypocalciuria, alkalosis, and all the clini- whitish flecking, and frequent caries, is the clas-
cal signs and symptoms of hypercalcemia. sic phenotype. Subjects are hyperactive, with
Chronic hyperphosphatemia will cause second- what is described as a “cocktail party patter” per-
ary hyperparathyroidism. sonality. They appear brighter than their full IQ
scores would suggest, as they have a great dis-
Granulomatous Disease crepancy between their verbal and mathematics
Disorders that demonstrate increased performance on testing. They are said to have
1α-hydroxylation of 25(OH)vitamin D which increased musical abilities on many instruments.
forms increased 1,25(OH)2D include granuloma- About 20 % of these children have hypercalce-
tous disease (sarcoid, eosinophilic granuloma) mia, hypercalciuria, nephrocalcinosis, and even
or various infectious causing granulomas renal impairment; the cause appears related to
including tuberculosis, histoplasmosis, coccidioi- abnormalities of the Williams syndrome tran-
domycosis, leprosy, or HIV and neoplasms scription factor (WSTF) encoded by BAZ1B
(lymphomas, dysgerminomas). Enzyme activity (*605681 BROMODOMAIN ADJACENT TO
is reportedly increased in monocytes in these ZINC FINGER DOMAIN, 1B; BAZ1B) which
conditions. Affected patients can experience leads to increased production and decreased deg-
hyperabsorption of dietary calcium and subse- radation of calcitriol. A low-calcium diet or the
quent hypercalcemia. Sarcoid patients also may use of glucocorticoids to block intestinal calcium
become hypercalcemic with small doses of vita- transport may be indicated in those affected with
min D. With increased sun exposure, hypercalce- hypercalcemia, but calcium levels tend to
mia is worse in sarcoidosis as serum 25(OH)D normalize with the passage of a few years.
increases. Glucocorticoid treatment may be used Idiopathic hypercalcemia of infancy (143880
to reduce intestinal calcium absorption, reduce HYPERCALCEMIA, IDIOPATHIC, OF
serum calcium values, and decrease 1,25(OH)2D INFANCY) is a rare and usually self-limited con-
values. dition that may represent a component of Williams
Hypercalcemia can also be found in cases of syndrome. Usually normal serum 1,25(OH)2D
inflammatory bowel disease. and 25(OH)D values are found. A low-calcium
152 7 Disorders of Calcium Metabolism and Bone

diet or glucocorticoid treatment may be pre- phosphate are administered during therapy. Oral
scribed, but most children are asymptomatic. phosphate may be administered to bind calcium
Fat necrosis with local dystrophic calcifica- in the intestine, but may result in diarrhea.
tion occurs in sick term or preterm infants who Bisphosphonates (described below) may be used
have had trauma, cold exposure, or hypoxia. Fat to lower serum calcium and a dose of 0.5–1 mg/
necrosis may lead to hypercalcemia due to the kg/dose by intravenous infusion over 4–6 h with
1-hydroxylase activity of macrophages and can careful monitoring of serum calcium. Calcitonin
persist for several days to weeks, but generally (2–4 U/kg/injection every 6–12 h) may be used to
resolves sooner without sequelae. Fat necrosis lower serum calcium but is not a long-term agent
may occur later in life as well. since tachyphylaxis develops in a few days. Oral
glucocorticoids decrease calcium absorption
Other Causes of Hypercalcemia from the intestines and may be used in severe
Various endocrine causes of hypercalcemia include cases of increased vitamin D or 1,25-dihy-
hyperthyroidism and glucocorticoid deficiency. droxyvitamin D but will not be helpful in hyper-
While pheochromocytoma can occur along with parathyroidism. Glucocorticoids also increase
hyperparathyroidism in multiple endocrine neo- urinary calcium excretion in addition to increased
plasia type 2A, 2B, pheochromocytoma by itself uric acid excretion so glucocorticoid therapy can
can produce parathyroid hormone-related protein lead to nephrolithiasis and nephrocalcinosis.
which may lead in rare cases to hypercalcemia. Ketoconazole is used as a therapy for fungal dis-
Thiazide diuretics limit the excretion of calcium ease but inhibits the formation of 1,25-dihy-
in the urine and thereby can raise calcium levels and droxyvitamin D as well as general steroid
serum. Hypercalcemia can result from hypervita- formation; if this agent is used for states of exces-
minosis A or the use of retinoic acid. Treatment sive calcitriol formation, careful monitoring for
with lithium can increase parathyroid hormone to a adrenal insufficiency is essential. Dialysis is
small degree leading to hypercalcemia. sometimes invoked in very severe hypercalce-
mia. Of course treatment of surgically correct-
able conditions such as hyperparathyroidism or
Treatment of Hypercalcemia parathyroid adenoma is another step. (For a dis-
cussion of the emergency treatment of hypercal-
The therapy for nonsymptomatic hypercalcemia cemia, see Chap. 15.)
allows time for mild measures such as increasing Information on a treatment that should not be
hydration, decreasing calcium in oral intake, and used in childhood: [Phenylalkylamines (cinacal-
limiting vitamin D intake including multivita- cet) are calcimimetic agents which have been
mins. Acute severe hypercalcemia is treated first used in adults to stimulate the calcium-sensing
with hydration with sodium chloride-containing receptor and thereby decrease parathyroid hor-
fluid until fluid volume is restored and only then mone secretion. However, the FDA has elimi-
the use of a loop diuretic, furosemide (1 mg/kg/ nated the use of this substance in children pending
dose given slowly intravenously is one option), an investigation into its safety.]
which will increase urinary calcium excretion,
may be added. It is extremely important to deter-
mine the state of hydration before the use of the Evaluation of Hypercalcemia
loop diuretic as dehydration may precede the use
of the diuretic and the diuretic will simply worsen Family history is important to determine if multi-
it. Treatment of dehydration is essential in the ple endocrine neoplasia as or other genetic condi-
treatment of hypercalcemia. Likewise long-term tions such as FHH are considerations. Maternal
use of loop diuretics may lead to nephrocalcino- health, diet, and complications of birth are like-
sis. Thiazide is not used as a diuretic since it will wise important as noted above. Evaluation of
limit calcium excretion and increase serum cal- symptoms of hypercalcemia may be negative
cium. In addition, sodium sulfate and sodium in mild cases of hypercalcemia. Laboratory
Hypercalcemia 153

evaluation may require multiple measurements of relationship and can lead to increased prevalence
calcium and multiple determinations of parathy- of pathologic conditions such as slipped capital
roid hormone due to variation in laboratory tech- femoral epiphyses, Blount’s disease, and fractures.
niques and results. An elevated serum protein will Immobilization reduces the ability of bone to
increase total serum calcium without parathyroid reach normal density, and even the limited activity
disease. Hyperparathyroidism may cause skeletal of children in our modern society has an effect of
abnormalities even though calcium and parathy- decreasing bone strength.
roid hormone levels are still in normal range but Any hypogonadal condition will also decrease
are in inappropriate relation to each other. Urinary bone mineral acquisition and increase fracture
calcium excretion must be determined. Skeletal risk. Excess thyroxine or PTH administration
evaluation is performed to look for changes caused cause decreased bone density. Glucocorticoid
by hyperparathyroidism. Imaging of the parathy- treatment in excess of physiologic amounts can
roid glands is done with ultrasound, MRI or CT, or stimulate osteoclasts and inhibit osteoblasts. Any
sestamibi (methoxyisobutylisonitrile) scans which of the conditions mentioned in this text that lead
will indicate parathyroid gland activity. to those endocrine abnormalities must be consid-
ered a risk factor for diminished bone density.
Osteoporosis Likewise chronic diseases often associated with
A peak increment of bone mineral accrual occurs increased inflammation as well as leukemia and
in infancy and in late adolescence. The adolescent other cancers are associated with low bone density.
accrual occurs after peak growth rate, and it may Immobilization may lead to hypercalcemia but
be this imbalance of bone size to mineral content may also lead to decreased bone density.
that causes the increased prevalence of fractures Since much of the determination of bone den-
in close to 5 % of normal adolescent boys. A low sity is genetic, those with a genetic background
impact fracture (sometimes called a pathologic suggesting susceptibility to osteoporosis may
fracture) is defined as a fracture which occurs at a already have more fragile bones at the time of
vertical distance less than the standing height of puberty. Idiopathic juvenile osteoporosis is rare
the individual. While a low impact fracture might and may be isolated or found in autosomal reces-
occur in the long bones, spinal fractures are other sive pattern (259750 OSTEOPOROSIS,
indicators of pathologic fractures. JUVENILE). The condition appears a few years
Low bone mass is inherited in a genetic pattern; before pubertal development, but some subjects
40–60 % of bone mass is determined by heredity. appear to resolve with the progression of puberty.
However, children and adolescents can modify In affected children extremity fractures due to
their bone mass by a non-genetic manner. Activity osteoporosis will cause the expected local pain and
leading to weight bearing increases bone mass, swelling; vertebral fractures, in addition, will
and adequate calcium, phosphorus, and vitamin diminish height. This condition must be differenti-
D intake likewise improve bone health. ated from mild osteogenesis imperfect (see below).
Inadequate calcium and vitamin D intake is Newborns are susceptible to decreased bone
endemic in youth in the USA which increases the density based on gestational age and nutritional
likelihood of bone disease. Further, ingestion of status as well as the mothers’ nutritional status and
carbonated beverages limits calcium-containing especially her vitamin D stores. Three-fourths
dairy-product ingestion, limits calcium absorption of the bone calcium content is accrued in the last
because of the phosphate content of the beverages, trimester, so premature infants are deprived of
and at least in females causes decreased bone much of this mineral. Further, abnormal GI func-
mass. Smoking decreases bone mass. Anorexia tion and absorption due to many disorders will
nervosa has many effects not the least of which is limit calcium, phosphorus, and vitamin D intake.
decreased bone mineral density; the female ath- A limitation exists to the amount of calcium and
letic triad likewise encompasses this finding (see phosphorus that can be infused in total parenteral
Chap. 9). Obesity, while increasing weight bear- nutrition, leading to greater risk of decreased bone
ing, does not increase bone mass in the normal in subjects receiving such support.
154 7 Disorders of Calcium Metabolism and Bone

Many chronic diseases will decrease bone den- adjusted for young adults of 21 years. If the
sity leading to secondary osteoporosis. Celiac dis- T-score is reported and used for diagnosis, every
ease is quite common and affects growth as well young child will automatically have osteoporosis.
as bone density as will other types of gastrointesti- Many inappropriate referrals are made because
nal disease and renal disease. Celiac disease will of the use of the T-score rather than the Z-score.
impair absorption of vitamin D. In the adult terms indicating bone mineral con-
Bone density and mineralization are important tent are osteopenia (−1.1 to 2.4 SD below the
concepts in the evaluation of rickets and osteopo- mean) and osteoporosis (−2.5 SD or more)
rosis. Bone mineralization refers to the addition of which is a more severe decrease in mineral con-
mineral, calcium phosphorus, and other minerals tent raising the likelihood of pathologic fractures.
into the bone matrix. Bone density refers to the In childhood and adolescence the term osteopo-
ratio of bone mass to volume and is expressed as rosis might be used for a child with low bone
grams over centimeters cubed. Dual X-ray absorp- mass (−2 standard deviations for age, sexual mat-
tiometry (DEXA) measures areal density which is uration, and gender) who has had a low impact
not true density since it is expressed as grams per fracture but more appropriately the terms low
centimeter squared representing the area under bone density or mass are applied in pediatrics.
evaluation. Quantitative computed tomography There are other methods of determining bone
(QTC) measures actual volumetric bone density. density. Volumetric bone density can be evalu-
The terms relating to bone evaluation are BMDtotal ated by qCT techniques, but they require a higher
which is the mineral density within the bone under dose of radiation, the CT scanner necessary to
the periosteal envelope and/or articular surfaces, perform the test is not readily available, and there
BMDcompartment which is the mass of mineral are limited standards for pediatrics. This tech-
per unit volume of trabecular or cortical bone, and nique is used in research studies however. MRI is
BMDmaterial which is the degree of mineraliza- used in research studies and offers potential for
tion of the organic bone matrix. measuring volumetric bone density in differenti-
Bone density evaluations are generally carried ating from cortical to trabecular bone. Qualitative
out by DEXA scan in children as the radiation ultrasound is a technique that requires no ionized
exposure is quite low and the information can radiation, but standards are not readily available
be related to fracture risk. DEXA measures the for children nor are clinical correlations.
BMDtotal and bone mineral content (BMC), but Evaluation of the etiology of diminished bone
results are for areal rather than the volumetric density will start with family history of osteopo-
results and the results vary by bone size (thin rosis or pathologic fractures and continue with
versus thick bone structure). Determination of nutritional history of the child. Evaluation for
volumetric bone density (three-dimensional) is coexisting conditions such as anorexia nervosa or
superior to determination of areal bone density other chronic conditions is important. Physical
(only two-dimensional), as thicker bones have examination will include determination of height
higher calcium content but may not be any denser in standard deviations or percentiles for age,
than smaller bones. Areal bone density is not as growth rate, and nutritional status by BMI.
closely related to fracture risk as volumetric bone Evaluation for a chronic disease or congenital
density. Adjustment for bone size is accom- conditions is important. Laboratory evaluation
plished by expressing bone mineral apparent includes measurement of serum calcium and ion-
density (BMAD) or estimated volumetric bone ized calcium, phosphate, magnesium, alkaline
density. DEXA cannot differentiate trabecular phosphatase, PTH, 25(OH)vitamin D, 1,25(OH)2
versus cortical bone. Bone density that is deter- vitamin D, creatinine, and urinary calcium and
mined by DEXA scan in children should be creatinine. In appropriate cases based on history,
expressed in Z-scores which are age and gender serum thyroxine and glucocorticoids and gonadal
adjusted (and ethnic group standards are also steroids are useful. A reflexive panel for celiac
available) as opposed to T-scores which are disease may be indicated.
Hypercalcemia 155

Treatment includes ensuring that normal vita- scarce and the agents have not been used for very
min D levels and intake are present; that calcium long. Therefore, it is recommended to perform any
intake is at least at the recommended level for age, necessary dental work before the use of bisphos-
if not more than recommended for basal levels; phonates if possible, and of course prevention of
and that physical activity is fostered as tolerated. dental disease is essential if the use of bisphospho-
Oral calcium citrate is a useful form of calcium nates is being considered. In view of the long-term
treatment or replacement; foods containing cal- side effects of bisphosphonates in the long term, as
cium in appreciable degrees include dairy prod- they are stored in the bone for many years, they are
ucts (cheese, yogurt, milk); sardines; dark leafy best used under the guidance of clinical studies in
greens including spinach, kale, turnips, and col- experienced centers.
lard greens; cereals fortified with calcium includ-
ing Total, Raisin Bran, and Corn Flakes; soy milk Osteopenia of the Premature Infant
fortified with calcium (which does not include all Since the fetus requires calcium and phosphorus
soy milk); and breads, grains, and waffles enriched during pregnancy, premature infants do not
with calcium. Precipitating factors such as gluco- receive this supply of the minerals necessary for
corticoid therapy must be minimized in an attempt bone development. Even recognizing this fact, it
to balance the beneficial effects of these medica- has been difficult to supplement premature
tions on the disease under treatment with the infants with enough calcium and phosphorus to
adverse effects of the medication on bones. equal the supply they would have received in
There are no medications approved in child- utero. Some medications for use in the premature
hood for the treatment of low bone mass. infant also interfere with a bone development.
Bisphosphonates are agents used in adults to Metabolic bone disease of prematurity is the term
impede osteoclast function and inhibit bone used to indicate these problems in premature
resorption but are not approved for children. While infants which may progress to the development
they have been successfully used in osteogenesis of rickets. Risks of fractures are high as 30 % of
imperfecta, there are reports of successful treat- premature infants weighing less than 1,500 g suf-
ment in osteopenia in selected children with fer fractures. Elevated alkaline phosphatase and
increased bone resorption. After the first use of decreasing phosphorous have generally been
bisphosphonates an acute phase reaction occurs used as an indicator of risk of osteopenia and
which appears to be an episode of infection with rickets in the premature infant.
vomiting and possibly a rash. Treatment leads to Long-term parenteral nutrition can lead to
higher density of the bone that is improved by the metabolic bone disease leading to osteopenia
treatment interspersed with lower density of the and progressive liver disease while leading to
osteoporotic bone leading to a fragility at these jaundice and failure to thrive. Elevated alkaline
junction sites which may lead to fracture. Some phosphatase in these conditions is frequently
suggest that the agents be used regularly once related to the bone disease.
started until bone growth ceases. There is hypocal-
cemia initially which leads to rebound hyperpara- Osteogenesis Imperfecta
thyroidism and increasing 1,25-dihydroxyvitamin Osteogenesis imperfecta is a group of condi-
D. It is generally suggested that the patient be tions characterized by increased fragility of
admitted for the 3 days of treatment for close bones and multiple fractures. They range in
observation but many are treated in the outpatient severity from mild to fatal. At the time of this
setting. Zoledronate is an agent offering a simpler writing there are 15 variations of osteogenesis
plan of treatment and is gaining favor over pami- imperfecta listed on Online Mendelian
dronate in children. There are reports of bisphos- Inheritance in Man. The numbering system is
phonate-related osteonecrosis of the jaw or based upon the date of the first report of the con-
BRONJ in adults, but the most recent review dition; this discussion will focus on the first
reports none in children. Nonetheless data are three types as examples of the conditions which
156 7 Disorders of Calcium Metabolism and Bone

are inherited in an autosomal dominant pattern. dense skull; narrowness of neural and vascular
Osteogenesis imperfecta type 1 (#166200 foramina; sandwich appearance of vertebral bod-
Osteogenesis imperfecta, type I) presents with ies; coxa vara; splayed metaphyses; hydrocepha-
relatively mild osteopenia with a variable degree lus; seizures (tetany); cranial nerve palsies;
of fractures which usually improves at puberty. pancytopenia; anemia; and extramedullary hema-
Wormian bones are intra-sutural bones found in topoiesis. Autosomal dominant osteoporosis
the skull sutures and are present in this condi- (#166600 OSTEOPETROSIS, AUTOSOMAL
tion. Blue sclera, thin skin, and easy bruising are DOMINANT 2; OPTA2) is due to a mutation in
characteristics on physical examination with the the chloride channel 7 gene (CLCN7). Bone mar-
stature lower than expected for the family. Mitral row transplants have been helpful in some forms
valve prolapse is also found. This condition may of osteopetrosis, and calcitonin has also been used
be confused with idiopathic juvenile osteoporo- with some success.
sis. Osteogenesis imperfecta type II (#166210
OSTEOGENESIS IMPERFECTA, TYPE II) is Nonaccidental Bone Injuries
extremely severe usually leading to perinatal The question may arise as to whether a fracture is
death; the child has fractures present at birth. nonaccidental or whether it is caused by osteopo-
Blue sclera and wormian bones are present. The rosis, osteogenesis imperfecta, rickets, hypo-
condition is due to mutation in the collagen I, phosphatasia, or other types of metabolic bone
alpha-1 polypeptide gene. Osteogenesis imper- disease in forensic cases. Due to the serious legal
fecta type III (#259420 ICD+OSTEOGENESIS implications in the consideration of nonacciden-
IMPERFECTA, TYPE III) also presents with tal injuries, it is best to involve an expert in the
prenatal fractures with more severe fractures field in a discussion with the child’s provider and
continuing after birth; the blue sclera noted at radiologist. Many cases arise before the child is
birth turns white with age. ambulatory which raises index of suspicion of
how a fracture can occur when the child has little
Increased Bone Mass Osteopetrosis mobility. Often a skeletal survey will be carried
Decreased osteoclast function results in a group of out to look for nonapparent fractures or meta-
eight congenital disorders known as osteopetrosis. bolic disease. Sometimes a skeletal survey or a
Autosomal recessive osteopetrosis (#259700 specific radiograph will be repeated in a few
OSTEOPETROSIS, AUTOSOMAL RECESSIVE weeks to see if any evidence of healing might
1; OPTB1) also called malignant form of osteopo- occur suggesting the recency of the event.
rosis is due to homozygous or compound hetero- Nuclear imaging of the bones is sometimes car-
zygous mutation in the osteoclast-specific ried out because there is increased specificity for
TCIRG1 subunit of the vacuolar proton fractures with this method than with a skeletal
pump while (#259710 OSTEOPETROSIS, survey. This is a situation where an expert opin-
AUTOSOMAL RECESSIVE 2; OPTB2) is ion is necessary.
caused by a mutation in the TNFSF11 gene.
Physical findings listed on OMIM involve multi-
ple organs and relate to the effects of increased Aluminum Toxicity
bone mass on skeletal integrity and stability func-
tion of nerves or bone marrow trapped within the Aluminum (Al) is found in parenteral nutrition
increasing bone mass. Thus, physical findings components including additives for calcium and
include frontal bossing; facial paralysis; deafness; phosphate, and premature infants are particularly
blindness; extraocular muscle paralysis; nystag- liable to toxicity. While the FDA recommends a
mus; optic atrophy; dental caries; distorted pri- maximum exposure of 5 μg/kg of aluminum, the
mary molars; hepatomegaly; splenomegaly; date of manufacturer of the nutrition formula related
uniformly dense skeleton; pathologic fractures; to the data of administration may cause significant
bone-within-bone radiographic appearance; thick, changes in content leading to excessive dosing.
Suggested Readings 157

Aluminum toxicity leads to central nervous system 12. Bhatt RN, Hibbert SA, Munns CF. The use of bisphos-
phonates in children: review of the literature and
changes and decreased mineralization of bone as
guidelines for dental management. Aust Dent
well as liver damage. J. 2014;59(1):9–19.
13. Misra M, Pacaud D, Petryk A, Collett-Solberg PF,
Kappy M, Drug and Therapeutics Committee of the
Lawson Wilkins Pediatric Endocrine Society. Vitamin D
Suggested Readings deficiency in children and its management: review of
current knowledge and recommendations. Pediatrics.
1. Shaw N. A practical approach to hypocalcaemia in 2008;122(2):398–417.
children. Endocr Dev. 2009;16:73–92. 14. Abrams SA, Committee on Nutrition. Calcium and
2. Root AW, Diamond Jr FB. Chapter 18—Disorders of vitamin D requirements of enterally a preterm infants.
mineral homeostasis in children and adolescents. In: Pediatrics. 2013;131(5):e1676–83.
Sperling MA, editor. Pediatric endocrinology. 4th ed. 15. Ross AC, Manson JE, Abrams SA, Aloia JF, Brannon
Philadelphia, PA: Elsevier; 2014. p. 734–845. PM, Clinton SK, Durazo-Arvizu RA, Gallagher JC,
3. Root AW. Chapter 8—Disorders of calcium and phos- Gallo RL, Jones G, Kovacs CS, Mayne ST, Rosen CJ,
phorus homeostasis in the newborn and infant. In: Shapses SA. The 2011 report on dietary reference
Sperling MA, editor. Pediatric endocrinology. 4th ed. intakes for calcium and vitamin D from the Institute
Philadelphia, PA: Elsevier; 2014. p. 209–276. of Medicine: what clinicians need to know. J Clin
4. Lips P. Vitamin D, physiology. Prog Biophys Mol Endocrinol Metab. 2011;96(1):53–8.
Biol. 2006;92(1):4–8. 16. Lui JC, Nilsson O, Baron J. Recent research on the
5. Allgrove J. Physiology of calcium, phosphate and growth plate: recent insights into the regulation of the
magnesium. Endocr Dev. 2009;16:8–31. growth plate. J Mol Endocrinol. 2014;53(1):T1–9.
6. Allgrove J. A practical approach to rickets. Endocr 17. Wit JM, Camacho-Hubner C. Endocrine regulation
Dev. 2009;16:115–32. of longitudinal bone growth. Endocr Dev. 2011;
7. Linglart A, Biosse-Duplan M, Briot K, Chaussain C, 21:30–41.
Esterle L, Guillaume-Czitrom S, Kamenicky P, 18. Misra M. Vitamin D insufficiency and deficiency in
Nevoux J, Prie D, Rothenbuhler A, Wicart P, Harvengt children and adolescents. UpToDate; 2014
P. Therapeutic management of hypophosphatemic 19. Carpenter T. Overview of rickets in children.
rickets from infancy to adulthood. Endocr Connect. UpToDate; 2014.
2014;3(1):R13–30. 20. Carpenter T. Overview of rickets in children. http://
8. Winzenberg T, Jones G. Vitamin D and bone health in www.uptodate.com/contents/overview-of-rickets-
childhood and adolescence. Calcif Tissue Int. in-children.
2013;92(2):140–50. 21. Carpenter T. Treatment of calcipenic rickets in children.
9. Crabtree N, Ward K. Bone densitometry: current status http://www.uptodate.com/contents/etiology-and-
and future perspectives. Endocr Dev. 2009;16:58–72. treatment-of-calcipenic-rickets-in-children.
10. Bergwitz C, Juppner H. Disorders of phosphate 22. Scheinman SJ, Drezner MK. Hereditary hypophos-
homeostasis and tissue mineralisation. Endocr Dev. phatemic rickets and tumor-induced osteomalacia.
2009;16:133–56. http://www.uptodate.com/contents/hereditary-
11. Davies JH. A practical approach to problems of hypophosphatemic-rickets-and-tumor-induced-osteo-
hypercalcaemia. Endocr Dev. 2009;16:93–114. malacia.
Disorders of Sexual Differentiation
8

The appearance of ambiguous genitalia in a new- which determines social sexual role. If a gene
born can present a biological and psychological mutation or exposure to an endocrine disrupting
emergency: the former because of possible electro- substance occurs, this normal progression may
lyte and glucocorticoid abnormalities and the latter not occur, and a disorder of sexual differentiation
because of the distress the parents will experience. (DSD) condition will lead to the phenotype of
This is a condition in which experience and careful ambiguous genitalia.
consideration are essential; immediate consultation
with a pediatric endocrinologist at the least or better
yet consultation with a team established for the eval- Genotype
uation and treatment of a child with a disorder of
sexual differentiation (usually composed of endo- Genotype or chromosomal sex is 46, XY for a
crinologist, geneticist, psychologist, surgeon/urolo- male and 46, XX for a female (Fig. 8.1). The Lyon
gist, and ethicist) is recommended before making a hypothesis stated that one of the X chromosomes
diagnosis, assigning sex of rearing, or embarking in a female is inactivated so that only one dose of
upon a treatment plan. Much has changed in the each gene on the X chromosome is active. It is
management of this condition with recent insights now established that inactivation does not occur
derived from long-term patient observations. on the entire X chromosome, as some genes
Indeed, the classifications of intersex conditions remain active and are required for normal sexual
presented in this chapter were only developed differentiation. This “relatively” inactivated X
approximately a decade ago with the input of chromosome appears as a Barr body at the periph-
patients and families as well as providers. These ery of the nuclear envelope on a Giemsa-stained
new terms replace the terms that will be found in preparation of cells scraped from the buccal
older textbooks which may lead to some confusion. mucosa. Patients will have one fewer inactivated
In this chapter, we will use the new terminology X chromosome (and one fewer Barr body) than
while on occasion referring to the old ones in case the number of X chromosomes, so that those with
the reader needs to cross-reference older articles. more than two X chromosomes will have more
than one Barr body (e.g., a patient with 48XXXY
will have two Barr bodies) and those with only
Normal Sexual Differentiation one X chromosome (45X Turner syndrome) will
have no Barr body. You may encounter Barr bod-
Normal sexual differentiation is based on geno- ies mentioned in the literature (and that is why it
type, which will determine gonadal sex, which is included in this chapter); we do not recommend
leads to phenotypic sexual differentiation, and this test be performed for clinical diagnosis as few

© Springer International Publishing Switzerland 2016 159


D.M. Styne, Pediatric Endocrinology, DOI 10.1007/978-3-319-18371-8_8
160 8 Disorders of Sexual Differentiation

Fig. 8.1 Genes found on the X chromosome of importance to sexual differentiation. From Grumbach and Conte in
Williams textbook of endocrinology 10th edition 2002. Used with permission

laboratories are capable of staining buccal smears meiotic recombination with the corresponding dis-
in a reliable manner and few physicians have tal pseudoautosomal areas of the X chromosome.
enough experience to interpret the buccal smear The SRY gene is located proximal to the boundary
results appropriately. Thus, a karyotype determi- of the pseudoautosomal region of Y chromosome.
nation is the only accurate method to determining The translocation of the SRY to an X chromosome
chromosomal sex. However, the determination of can cause an XX individual, who inherited one
chromosomal sex does not itself determine the normal X chromosome from the mother and one X
management of the child as many more consider- chromosome which now has an SRY gene from
ations must be taken into consideration. the father, to develop testicular tissue; this is the
The Y chromosome contains the SRY gene, origin of an XX male, an individual who would
which directs the differentiation of the bipotential demonstrate many characteristics that would be
gonad in the early fetus into a testis. Active genes found in a 47XXY male with Klinefelter’s syn-
on portions of the distal arms of the Y chromo- drome. The SRY also can be transferred to an auto-
somes, the pseudoautosomal regions, can undergo some, allowing an XX phenotypic male to result
Normal Sexual Differentiation 161

Fig. 8.2 The major


steps and genes and
INTERMEDIATE
hormones involved in
MESODERM
gonadal sexual
differentiation SF-1
WT-1

PLACENTA
UROGENITAL
RIDGE
Y chromosome X chromosome
PAR SOX9

SRY
BIPOTENTIAL
GONAD

Duplication of Ovarian
DAX-1 or WNT-4 genes

SOX9
hCG WNT-4
Leydig SOX9 OVARY
cells
TESTIS
FEMALE SEXUAL
Sertoli Steroidogenic DEVELOPMENT
cells genes
SF-1 SOX-9
WT-1 AMH gene SF-1

Testosterone Support of
AMH wolffian ducts
5 Androgen
AMH Receptor reductase receptors

Dihydrotestosterone Masculinization
of external
Regression of genitalia
MuIlerian ducts
MALE SEXUAL DEVELOPMENT

from this anomaly. Alternatively, 46, XY pheno- so that ovarian development is no longer consid-
typic females lack testicular development because ered a passive process and the development of a
of the loss of SRY from the Y chromosome inher- testis an active process as was the case in the
ited from the father. Other autosomal genes deter- past. Abnormalities in the genes causing testicu-
mine sexual differentiation; abnormalities of these lar development in a 46, XY individual character-
regions also may lead to disorders of sexual dif- istically lead to ambiguous genitalia or sex
ferentiation (Fig. 8.2). reversal, while abnormalities in the genes caus-
ing ovarian development of a 46, XX individual
usually manifest later in life as absent or delayed
Gonadal Sex menarche or premature menopause. Dysgenetic
testicular development leads to a risk for neopla-
Gonadal sex is determined by the presence or sia and might be found, for example, in an indi-
absence of the SRY. There are genes that cause vidual with an apparent 45X karyotype with
the indifferent gonad to develop into an ovary in mosaicism for a small number of 46, XY cells.
the absence of SRY, including Wnt4 and Bmp2, Fluorescence in situ hybridization probes for the
162 8 Disorders of Sexual Differentiation

Y chromosome are used to determine if an indi- development of these two areas is based on the
vidual has a Y cell line indicating the presence of effect, or lack thereof, of androgens and proteins.
dysgenetic testicular tissue that may become neo- Internal sexual ducts (Fig. 8.4) start as bipo-
plastic even though the gonad was expected to be tential, but one set develops and the other degen-
a streak gonad comprised only of fibrous tissue. erates as sex is determined. Internal genital ducts
If there is only one X chromosome and no SRY, are masculine if derived from the Wolffian ducts
the gonad will develop into an ovary during the or feminine if derived from the Müllerian ducts.
fetal period and the ovary will contain ova. The In the presence of the testicular peptide product
ova, however, will atrophy in the absence of a derived from the Sertoli cells, anti-Müllerian
second normal X chromosome, and the ovary hormone (AMH or MIF for Mullerian Duct
will degenerate into the “streak gonad” character- Inhibitory Factor), a member of the TGF-beta
istic of Turner syndrome. Many other genes are superfamily, the Müllerian ducts will atrophy
involved in the creation of the gonad, so this through paracrine action of AMH; in the absence
schema must be considered a simple summary. of AMH, the uterus, oviducts, and the upper two-
The steroid metabolism of the gonad involves thirds of the vagina will develop spontaneously
several enzymes in the conversion of cholesterol into a basic form. Gonadal development in 46, XY
into androgens or estrogens; many of these enzymes DSD (previously termed male pseudohermaphro-
also are important in the biosynthetic activities of ditism) may lead to more Leydig cell function
the adrenal gland, so that a genetic defect in an and testosterone production than AMH produc-
enzyme may affect both gonadal and adrenal func- tion, and some level of Müllerian development
tion. Figure 8.3 demonstrates the steroid molecules, may occur in a genetic male with impaired AMH
the enzymes involved and the names of the genes production due to dysgenetic testes in these
that identify them. Steroid determinations and cases. In the presence of high local concentra-
genetic probes in clinical or research laboratories tions of testosterone, the Wolffian ducts will
will detect these enzymatic defects. develop into the rete testes, efferent ducts, epi-
Primordial germ cells develop in the primary didymis, vas deferens, and seminal vesicles
ectoderm and migrate to the dorsal wall of the through paracrine action of the testosterone. As
yolk sac. By the fifth to sixth week the primordial the testosterone and AMH will come from a tes-
germ cells migrate to the midgut and hind gut. By tis, a unilateral testis in ovotesticular DSD (previ-
the seventh and eighth week they have migrated ously termed true hermaphroditism) could
through the dorsal mesentery into the gonadal produce Müllerian regression and Wolffian dif-
ridges formed near the nephrotic cord where by ferentiation on the ipsilateral side, whereas a con-
the ninth week they colonize their respective tes- tralateral ovary could allow the development of
tes or ovaries. Thereafter with appropriate genetic contralateral oviduct and a hemiuterus. Thus in a
composition, they will differentiate into oogonia ovotesticular DSD, one side of the internal sexual
in the fetal ovary or spermatocytes in the fetal tes- ducts may appear similar to male ducts and the
tis. Meiosis is the process of cell division in germ other to female ducts.
cells that leads to the development of haploid External genitalia (Fig. 8.5) are bipotential
cells which can become oogonia or spermato- until 8 weeks of gestation. Without the presence
cytes. Germ cells in the ovary immediately begin of characteristic male concentrations of andro-
meiosis as they develop into oogonia, but germ gens, the genital tubercle will become a clitoris,
cells in the testes become quiescent until meiosis the urogenital folds will become the labia minora,
begins in the peripubertal period. and the labia majora will form from the labio-
scrotal swelling. In the presence of systemic cir-
culating testosterone, which is locally converted
Phenotypic Sex into dihydrotestosterone (DHT or 5a-androstane-
17b-ol-3-one) by the 5α-reductase enzyme in the
Phenotypic sexual development involves the inter- sexual skin, and in the presence of normal andro-
nal ducts and external genital appearance. The gen receptors (ARs), a boy will be virilized into a
Normal Sexual Differentiation 163

Fig. 8.3 Normal steroid


physiology in the adrenal
gland and the gonads as
described in the text.
Enzymes are noted in blue;
major products are noted in
red. The pathways most
prominent in the gonads are
on the right with some of the
steps also found in the zona
fasciculata of the adrenal
gland

classic male appearance. With DHT, the genital for male development, as fetal gonadotropins are
tubercle will differentiate into a penis, the uro- not yet secreted from the fetal pituitary gland in
genital slit will form a penile urethra, and fusion substantial amounts. During the last two trimes-
of the labioscrotal folds will produce a scrotum. ters as hCG wanes, fetal pituitary LH is secreted
The fetal testes must be functional to allow in the greater amounts that are necessary to stim-
these steps of virilization to progress. In the first ulate testicular activity and cause further normal
trimester, maternal human chorionic gonadotro- growth of the penis; if the fetal pituitary gland
pin (hCG) stimulates the LH/hCG receptor on the cannot produce luteinizing hormone (LH), the
fetal testes to produce the testosterone necessary penis will be normally shaped (because of early
164 8 Disorders of Sexual Differentiation

Fig. 8.4 A schematic depiction of the development of the internal sexual ducts when testosterone exerts or does not
exert its normal effects (Adapted from an original figure of Dr. J. Wilson with permission)

hCG stimulation of the fetal testosterone produc- female is exposed to the androgen after 8 weeks
tion) but small because of lack of the enlargement and before 13 weeks of gestation, the vaginal
that only occurs if stimulated by fetal LH; with- opening may fuse posteriorly to become slit-
out fetal LH the penile length will likely be less like, and the urogenital slit may enclose at least
than 2.0 cm (which is −2.5 SD from the mean) part of the urethra; androgen exposure after 13
and is called a microphallus, as compared with an weeks of gestation will only enlarge the length
average normal length of 3.5 cm at term. and thickness of the phallic structure, causing
Some of the enzymes involved in the produc- clitoromegaly without affecting the vaginal
tion of sex steroids on the same as found in the opening. Average term penile size is 3.5 (gently
adrenal gland as described in Chap. 10. The zona stretched) × 1 cm and clitoral size is about
fasciculata of the adrenal gland produces sex ste- 4 mm × 3 mm. Thus, the appearance of the geni-
roids, but some of the steps are only found or talia at birth reveals much about the physiology
more active in the testes and ovaries (Fig. 8.3). of the fetus.
Disorders of the normal steroid biosynthetic The anogenital distance measures from the pos-
pathway are described below. terior of the scrotum of the male or of the posterior
Any interruption of the normal progression fourchette of the female to the center of the anus.
toward masculine development in a 46, XY The distance is related to androgen exposure as the
fetus will result in the development of some distance is longer in males (19.81/−6.1 mm) than
degree of ambiguous genitalia. Any exposure to in females (9.11/−2.8 mm). Lengthening of the
androgens at a sensitive time in development anogenital distance in girls occurs with excessive
will cause a 46, XX fetus to virilize to some androgen exposure, and shortening of the anogeni-
degree also causing ambiguous genitalia. If the tal distance in boys occurs with inadequate andro-
Normal Sexual Differentiation 165

Fig. 8.5 A schematic depiction


of the changes to the external
genitalia of the developing fetus Genital
when testosterone is converted to fold
DHT by interaction with the Genital
androgen receptor. On the left in swelling
a normal male. Male develop- INDIFFERENT
ment can only proceed with the Genital STAGES
presence of adequate amounts of tubercle
dihydrotestosterone (DHT) and
normal androgen receptors.
Female development in the
absence of DHT is presented on
the right (Adapted from an MALE FEMALE
original figure by Dr. J. Wilson
with permission)
Glans Glans

Urethral
Fused groove
genital folds
Anus Anus
Urethral
groove
Labia minora
Labia majora
Scrotum

Prepuce

Body of
penis Clitoris
Urethral oritice
Scrotal Hymen
raphe

gen production or in the presence of endocrine of gender of sexual partners; and (d) the desire
disruptors which lessen androgen effects. and capacity for parenting.
The classic view has been that gender identity
is determined mainly by the surroundings of the
Social Gender growing child, but because the physical appear-
ance of the child influences those interacting with
Gender development may be thought to consist the child, gender identity is also affected by phys-
of (a) gender identity, the experience of one’s self ical sexual differentiation. Interactions with fam-
as female or male; (b) gender role, the behavior ily and peers will teach the child how to act within
that identifies to others or self whether one is the expected gender role. However, a growing
male or female; (c) sexual orientation, the choice body of research demonstrates effects of prenatal
166 8 Disorders of Sexual Differentiation

androgen exposure on subsequent gender devel- 46, XX DSD: Conditions Causing


opment, although human beings retain plasticity Genetic Females to Become Virilized
to allow some alteration to the effects of prenatal (Previously Called Female
androgen exposures. For example, a girl exposed Pseudohermaphroditism)
to prenatal androgens, because of virilizing CAH,
if raised socially as a girl, will have a normal These 46, XX patients are exposed to excessive
female gender identity but may demonstrate some amounts of androgen which can be converted to
male tendencies in play behavior and reasoning DHT during fetal life causing virilization of
and/or a male tendency in gender role. Conversely, external genitalia. In the absence of anti-
males who are exposed to prenatal androgens but Müllerian hormone (since there is no testis nor
have inadequate male external genitalia and are Sertoli cells), internal ducts will be derived from
therefore raised as females may choose a male Müllerian ducts and will be female ducts includ-
gender identity spontaneously in later childhood ing ovaducts and uterus leading to the possibility
or adolescence. Thus, in cases of 46, XY DSD, of fertility.
described later, decisions on sex of rearing are Congenital adrenal hyperplasia is detailed
difficult, and recommendations are presently con- below since the most common ones present with
troversial and undergoing reconsideration. Patient ambiguous genitalia. The basis of steroidogene-
groups have been forceful in suggesting that gen- sis is discussed in Chap. 10, and aspects of adre-
der assignment and surgical reconstruction should nal insufficiency are found there as well.
await such time as the child can decide; others
believe that this is unworkable, and gender should Virilizing Congenital Adrenal
be assigned in an unambiguous manner early. All Hyperplasia
would agree that parents must be involved and An abnormality in any of the enzymes involved in
that the best understanding of the biology of the the production of cortisol will, in the absence of
condition must be considered. It has been consid- feedback inhibition, allow increased adrenocorti-
ered difficult to institute change of gender suc- cotropic hormone (ACTH) secretion. The adrenal
cessfully after age 18 months (except for the case glands, unable to produce the glucocorticoid end
of 5α-reductase deficiency discussed later), and product, cortisol, which would ordinarily suppress
concern remains over the psychological effects of ACTH, will become hyperplastic. Depending on
spontaneous or guided change in gender identity the enzyme affected, the adrenal gland may make
later in life. Legal issues arise in considerations of extremely increased androgens which will cause
gonadectomy in affected children before the age virilization of a female fetus. Alternatively, a
of consent and advocacy groups have brought suit defective enzyme which is essential for testoster-
for their assertions that gonadectomy have been one production will cause inadequate virilization
improperly performed in young children. For all in a male. A defect of an enzyme in the pathway
of these complexities the DSD team described at involved in the production of aldosterone will
the beginning of this chapter, usually found in cause sodium loss and the retention of potassium,
children’s hospitals or university centers, should leading to hyponatremia and hyperkalemia, possi-
be consulted. Gonadectomy for other causes than bly to a life-threatening extent although some of
neoplasia should not occur without consideration these salt-losing conditions do not demonstrate
of the future social and legal considerations. ambiguous genitalia (see Table 8.1).

21-Hydroxylase Deficiency (*201910


Disorders of Sexual Differentiation ADRENAL HYPERPLASIA, CONGENITAL,
DUE TO 21-HYDROXYLASE DEFICIENCY)
The present term for what was previously called 21-Hydroxylase deficiency, the most common
an “intersex condition” is a disorder of sexual dif- type of CAH, is the most common cause of
ferentiation or DSD. ambiguous genitalia and virilization in a
Disorders of Sexual Differentiation 167

Table 8.1 Differential diagnosis of adrenal enzyme effects


Newborn Postnatal Salt losing=+
Deficiency Phenotype Virilization Hypertension=*
StAR (also known as lipoid Infantile female − +
congenital adrenal hyperplasia)
3β-Hydroxylase (3βHSD) Ambiguous in XY + +
Female or ambiguous in XX + +
17α-Hydroxylase (P450c17) Infantile female − *
11β-Hydroxylase (P450c11β) Male in XY + *
Ambiguous in XX + *
21-Hydroxylase (P450c21) Male in XY + +(50 %)
Ambiguous in XX + +(50 %)
18-Hydroxylase (P450c11AS) Normal − +

newborn female, and the nonclassic type of type 2 and aldo-keto reductase subfamily 1C and
21-hydroxylase deficiency is the most common subsequent conversion to androstanediol and
type of genetic disease in human beings. dihydrotestosterone by 17-beta hydroxysteroid
21-Hydroxylase deficiency CAH is best consid- dehydrogenase type VI was described. This new
ered as a spectrum of diseases ranging from the understanding suggested there may be better
mild nonclassic or cryptic form to the simple vir- markers for the control of congenital adrenal
ilizing form to the salt-wasting form. The hyperplasia than 17-hydroxyprogesterone and
21-hydroxylase enzyme is a P450-class enzyme that improved treatments may be developed. All
with the gene (denoted as cpy 21A2) located on patients with 21-hydroxylase deficiency have
chromosome 6 (6p21.1) in proximity to the elevated 17-hydroxyprogesterone at diagnosis in
human leukocyte antigen (HLA) locus adjacent the basal state in the congenital (severe) form or
to the genes for the fourth component of comple- after ACTH stimulation in the late-onset (non-
ment. Progesterone is 21-hydroxylated into classic or cryptic) form discussed later.
deoxycorticosterone (DOC) in the pathway Classical CAH condition occurs in about 1 in
toward the production of aldosterone and 15,000 births. About 50 % of patients with the
17-hydroxyprogesterone [17(OH)progesterone], congenital form have clinical salt loss and hyper-
or 17OHP is 21-hydroxylated to 11-deoxycorti- kalemia because of impaired production of
sol in the pathway toward cortisol. Thus because DOC. Within one family, if a proband has salt los-
the production of cortisol is diminished due to a ing, other affected children also will be salt losers;
defect in the 21-hydroxylation step, ACTH further, the affected children within one family
increases and stimulates the production of prod- will share the same HLA types, allowing a possi-
ucts of the adrenal gland proximal to this defect: bility of prenatal diagnosis of future pregnancies.
the main product is 17(OH)progesterone and its Females with 21-hydroxylase deficiency will be
precursors. Elevated 17-hydroxyprogesterone virilized in utero due to elevated androgens that
concentrations may be converted into testoster- reach the externa genitalia by classic endocrine
one by either of two metabolic pathways. In the mechanisms, demonstrating any variation of clitor-
classic pathway conversion of 17-hydroxypro- omegaly, posterior fusion of the labia majora some-
gesterone to androstenedione and testosterone, times into the appearance of an “empty scrotal
dihydrotestosterone occurs by 17,20 lyase sack,” and lack of formation of the vaginal vesicular
enzyme activity which is inefficient. Recently, septum, causing a urogenital sinus. Thus, a 46 XX
the more efficient “backdoor” pathway which female will have an appearance on the continuum
causes the conversion of 17-hydroxyprogester- from ambiguous genitalia to “male with unde-
one to androstenedione through 5 alpha reductase scended testes.” However, the internal ducts remain
168 8 Disorders of Sexual Differentiation

female in the absence of AMH or local paracrine appear. Thus, in these days of short-stay deliver-
exposure to high concentrations of androgens ies, a patient may actually be discharged from
which require a testes to be present and functional. the hospital before biochemical complications
Exposure to prenatal androgens characteristic of develop. The initial signs of electrolyte distur-
CAH is reported to exert an effect on behavior as bance may be vomiting and may appear as a
girls as those with CAH exhibiting more male like case of viral gastroenteritis on casual evaluation.
play and behavior than their unaffected sisters. A Boys who look normal may be mistakenly diag-
male infant with 21-hydroxylase deficiency will not nosed as having pyloric stenosis and even
appear obviously abnormal and may not be diag- undergo unnecessary GI surgery which may be
nosed until a metabolic emergency occurs, which is fatal due to untreated adrenal insufficiency; one
why newborn screening for 21-hydroxylase defi- diagnostic difference is that patients with
ciency is so important. On the contrary, a girl with 21-hydroxylase deficiency have high potassium
ambiguous genitalia will usually receive immediate concentrations and often acidosis, whereas those
attention because of more immediate diagnosis with pyloric stenosis have low potassium and
caused by the appearance of ambiguous genitalia. metabolic alkalosis. If untreated, salt loss can
Newborn screening programs in most lead to sodium values in the 110 mEq/L or lower
locations regularly include detection of range and elevated potassium to the range
17-hydroxyprogesterone values on their panel to greater than 9.5 mEq/L by age 3–4 weeks if the
ensure early treatment of 21-hydroxylase defi- child survives that long without therapy.
ciency. However, premature infants will not yet Vomiting can lead to weight loss, so that body
have appropriate development of the definitive weight decreases to a point lower than birth
zone of the adrenal gland so that 3-beta-hydroxys- weight. Simple virilized patients have no obvi-
teroid dehydrogenase and 11-beta-hydroxylase ous salt loss but may have more subtle decreases
activity are not active to the levels that occur at in total body sodium leading to elevated plasma
term, because premature infants do not have the renin activity (PRA), suggesting the need for
characteristic postnatal decrease in activity of the extra salt or mineralocorticoid therapy in spite
fetal zone of the adrenal gland. Thus premature of the absence of the clear diagnosis of salt-los-
infants may have a positive newborn screen or an ing adrenal hyperplasia.
indeterminate value of 17-hydroxyprogesterone Skin pigmentation may darken in flexural
leading to the necessity of secondary screening. If surfaces, on the areola, or generally, a band of
the child being evaluated is a premature infant pigment may occur at the gumline while the
with a normal female phenotype without clitoral patient is untreated because of extremely
enlargement nor partial fusion of the vaginal open- increased ACTH secretion, but this pigmenta-
ing, it is virtually impossible that the child would tion will not occur until after the immediate
have classic virilizing 21-hydroxylase deficiency, neonatal period. Androgen secretion will lead
thus indicating that the screen was a false positive. to peripheral sexual precocity in affected chil-
A normal-appearing phenotypic male however dren who survive the neonatal period in the
will have no visual signs to indicate whether the untreated state, with the appearance of pubic
positive newborn screen is a false-positive or a hair, clitoral or penile enlargement, acne, deep-
true diagnostic finding. Premature infants with ening of the voice, muscular development, and
false-positive neonatal screens for 21-hydroxylase rapid growth. Aromatization of the androgens
deficiency will with the passage of time develop to estrogens will advance bone age over the
normal enzyme function and the abnormal values months and years after birth. Therefore,
will decrease. although the child will be large for age, the
In the salt-losing form of 21-hydroxylase rapid bone-age advancement will later lead to
deficiency, salt wasting does not manifest until early epiphyseal fusion, and short adult stature
after several days of age. Likewise, hypoglyce- will result. The testes will not initially enlarge
mia does not always develop in the immediate in affected boys because they are not stimulated
neonatal period but may take a day or two to by gonadotropin secretion since the virilization
Disorders of Sexual Differentiation 169

is caused by the androgens and precursors of Jewish women, making this disorder the most
the adrenal gland. Elevated androgens will also common genetic disease in human beings.
cause maturation of the hypothalamic–pituitary– Nonclassical congenital adrenal hyperplasia
gonadal axis. When glucocorticoid treatment is presents in a very similar manner to PCOS as
administered and adrenal androgen secretion described in Chap. 9. The nonclassical congenital
decreases, the adrenal androgens that were sup- adrenal hyperplasia is a disorder solely of the
pressing gonadotropin secretion diminish, and adrenal glands and not the ovaries. Polycystic
true central precocious puberty may develop ovarian disease is mainly based in the ovaries,
(with pubertal testicular enlargement then and each condition requires different treatment.
developing in boys). This secondary complica- We will use the term cryptic 21-hydroxylase defi-
tion will further compromise adult height by ciency for patients with laboratory values identi-
causing bone-age advancement; central preco- cal to those found in patients with the late-onset
cious puberty after exposure to excess andro- form, but without virilization: cryptic 21-hydrox-
gens can be treated with gonadotropin-releasing ylase deficiency appears to be a variant of the
hormone (GnRH) analogs, as can any other late-onset form.
cause of central precocious puberty (see Chap. 9).
Testicular enlargement may also occur because Diagnoses
of enlargement of testicular adrenal rest tissue The diagnosis of 21-hydroxylase deficiency is made
(TART) which refers to embryonic adrenal tis- by the elevation of serum 17-hydroxyprogesterone
sue remaining in the testes which can be stimu- concentrations, the precursor of the enzyme that is
lated by ACTH. This condition previously was deficient, in the basal state or 60 min after 250 mg
thought to occur in poorly treated patients, but synthetic ACTH (1–24 ACTH or Cortrosyn) given
it is now apparent that all boys with congenital intravenously. Note that the low-dose ACTH stimu-
adrenal hyperplasia are susceptible to the devel- lation test mentioned in Chap. 10 is not appropriate
opment of TART. Long-term studies indicate for this diagnostic activity. Serum concentrations of
that TART tissue may become neoplastic, so 17-hydroxyprogesterone are elevated in cord blood
male patients should be followed up carefully. of normal infants compared to blood samples sev-
There are various terms used in the literature eral days later because of the activity of the fetal
for the mildest types of 21-hydroxylase defi- adrenal zone at that early stage as well as contribu-
ciency: nonclassical, cryptic, or late-onset tions from the mother’s steroid metabolism.
21-hydroxylase deficiency. We will use the term However, 17-hydroxyprogesterone values decrease
nonclassical 21-hydroxylase deficiency which is to levels of 100–200 ng/dL within 24 h after birth.
characterized by normal phenotype at birth, and A neonate with classic 21-hydroxylase deficiency
therefore this is not considered 46, XX DSD will have concentration of 17-hydroxyprogesterone
(i.e., no clitoromegaly nor posterior vaginal in the 5,000–10,000 ng/dL range or higher, allow-
fusion occurs in girls), with androgenic effects ing differentiation from normal infants. However,
only occurring years after birth, in childhood, due to widespread neonatal screening using sensi-
adolescence, or young adulthood. The serum tive techniques, some children with nonclassical
17-hydroxyprogesterone and other adrenal CAH may be discovered at birth. Methods of
androgen concentrations may be elevated in the screening for 21-hydroxylase deficiency in heel-
basal state but definitely will be elevated after stick samples taken after birth are instituted in the
ACTH stimulation, but they will be not as high as newborn screening programs of all states. As stated
found in the congenital form of CAH. Female above, false-positive screens are common in prema-
patients may complain of increasing facial or ture infants due to the immature state which allows
body hair, and menstrual abnormalities may continued fetal adrenal zone function for a period
occur. The incidence varies in ethnic groups, with after birth in contrast to term baby when the fetal
an incidence of 1:1000 in the general Caucasian adrenal zone rapidly decreases. Prenatal diagnosis
population and as much as 1:50–27 in Ashkenazi is available for 21-hydroxylase deficiency by mea-
170 8 Disorders of Sexual Differentiation

suring amniotic fluid steroid metabolite values or by considered normal on the basis of a 3-h collection
matching the HLA type of the fetus under consider- that was fallaciously low because of low volume
ation (taken from amniocentesis or chorionic villus) and inadequate time of collection. A 24-h urinary
to an affected proband sibling of known HLA type pregnanetriol determination, a reflection of
or by restriction fragment length polymorphism 17-hydroxyprogesterone secretion, has the same
analysis of fetal tissue. Recent techniques are devel- reliance on a full 24-h urine collection.
oped to allow diagnosis on the scarce fetal blood Diagnosis in the newborn rests upon analysis
cells that enter the maternal circulation. A mother of a serum sample for 17-hydroxyprogesterone
with a proband with 21-OH CAH who is pregnant as well as other metabolites of the adrenal gland;
again may be treated with glucocorticoids that pass laboratories experienced with pediatric endocrine
the placenta to decrease virilization of a potentially practice will treat the situation as a medical emer-
affected female fetus. Prenatal treatment involves gency and analyze the blood within a day or two.
the use of dexamethasone as cortisol and predniso- These experienced laboratories will make deter-
lone are deactivated by the placenta as they are con- mination on small samples of blood that are
verted to inactive cortisone and prednisone by 1 appropriate in a newborn rather than the large
beta HSD 2. samples requested when diagnosing a young
If the chorionic villus sample determines the adult with hirsutism. A panel of precursors to
fetus is male or if endocrine testing of amniotic various causes of intersex can be carried out on
fluid shows the fetus is not affected, glucocorti- very small samples in national laboratories with
coid therapy is stopped. Each parent is heterozy- pediatric standards. New highly sensitive analy-
gous for the CAH gene; there is a one in four ses of urinary metabolites of adrenal androgens
chance of an offspring having homozygous con- are becoming available on spot collections and
genital adrenal hyperplasia, and of those one will may further simplify diagnosis.
be a female who can be viralized; thus there is a As salt loss is not usually seen in a salt-
one in eight risk of a treatable prenatal condition. losing patient until after about age 5 days, vigi-
Thus, seven of eight fetuses are unnecessarily lance must not be relaxed until days to weeks of
treated with glucocorticoids for 8–12 weeks of carefully observing the patient’s electrolyte sta-
gestation and are subjected to potential risk from tus pass. Since hemolysis is possible from a
this treatment. Fetal treatment with glucocorticoid heelstick sample which will raise the potassium
administered to the mother may diminish viriliza- value, a carefully collected venous sample
tion of affected females but is still considered should be obtained if questions of hyperkale-
experimental therapy. While some reports suggest mia rise. A plasma renin activity (PRA) should
this is safe therapy, there are cautions as orofacial be obtained to confirm salt loss, but the result of
clefts, decreased birth weight, poorer verbal this test may not be available for several days,
working memory, and poorer self-perception and and immediate diagnosis cannot therefore rest
scholastic and social competence are also on this test. Plasma renin activity values must
reported, and it is cautioned that this treatment be be correlated with age in the laboratory with
carried out in controlled studies with experienced pediatric standards as the values are higher in
practitioners of the method if at all. children than adults.
The risk of cleft palate due to fetal glucocorti- If an ultrasound examination in a newborn
coid excess adds further caution when consider- with ambiguous genitalia reveals a uterus, the
ing this method. patient will most likely be 46, XX DSD and
Older methods of diagnosis, now superseded, therefore will most likely be a female with
include analysis of a 24-h urine collection for 21-hydroxylase deficiency based upon probabil-
17-ketosteroid as a reflection of adrenal androgen ity. However, it is essential to confirm the diagno-
secretion (primarily DHEA or dehydroepiandros- sis and not simply guess based on the prevalence
terone). Difficulties occur if a full 24-h collection of 21-hydroxylase CAH as other diagnoses
is not obtained; we have seen patients mistakenly remain possible. A vesicovaginogram with careful
Disorders of Sexual Differentiation 171

technique, using a rubber dam to contain the radi- growth, at the time of this writing this approach is
opaque dye, will reveal the anatomy of the not widely practiced in growing children and is
internal vaginal structures and whether absence not included in the current guidelines. However,
of the vesicovaginal septum is causing the devel- synthetic glucocorticoids such as prednisone,
opment of a urogenital sinus. prednisolone, or dexamethasone have their place
Radiological evidence in the evaluation of in the treatment of older individuals once growth
21-hydroxylase deficiency which is untreated for ceases. The dose of hydrocortisone is variable
a longer time will include an advanced bone age among individuals, but for an initial dose,
(a simple virilized child may survive this long if 13–15 mg/m2 or more of cortisol is useful (The
no serious stresses encountered, but a child who ultimate dose may be greater than the normal
is salt losing will become profoundly ill before secretory rate of cortisol, because the goal is not
this age). An enlarging pituitary contour (due to simply to replace the adrenal cortisol production
basophil hyperplasia) on MRI or erosion of the but also to suppress abnormal product formation,
sella turcica will develop lateral skull radiograph and adrenal production is elevated even more due
if the patient is un- or undertreated for years. to the adrenal hyperplasia). Once control occurs
Genetic testing for 21-hydroxylase deficiency and adrenal hyperplasia is reduced, lower doses
is available in commercial laboratories. The may be invoked based upon growth, steroid val-
functional gene for 21-hydroxylase is CYP 21A2 ues, and bone-age advancement in laboratory
but there is an inactive pseudo-gene, CYP 21 testing. Oral Cortef is usually administered in
A1P which is about 98 % identical to the func- doses every 8 h. The dose should be titrated to the
tional gene. 95 % of cases of 21-hydroxylase child as noted later. Some recommend that the
deficiency are accounted for by about 11 muta- greater portion of the dose be given at bedtime so
tions, and rather than sequencing the entire gene, the early-morning peak of ACTH will be sup-
targeted analysis of these mutations can be car- pressed and less androgen will be secreted, but
ried out. Genetic analysis is not usually necessary there is little evidence that one method is better
to identify an individual with 21-hydroxylase than the other (this is the reverse of the schedule
deficiency as biochemical testing will usually for the treatment of adrenal insufficiency without
suffice, but it is a means of determining whether CAH; see Chap. 10). There is no place for solu-
a fetus may be carrying the mutation present in tions of hydrocortisone made up by a compound-
an affected family once the proband has been ing pharmacy as the variation in the daily dose
identified. will be far too great to allow careful treatment. If
For the differential diagnosis of adrenal an emergency arises, intravenous or intramuscu-
enzyme defects, see Table 8.1. lar hydrocortisone hemisuccinate (Solu-Cortef)
is used. Intramuscular cortisone acetate takes
Treatment hours to initiate effect and is not an appropriate
Virilizing 21-hydroxylase deficiency is treated medication to use in an emergency.
with glucocorticoid replacement, and the most Many methods are proposed to best follow the
common approach is the use of natural com- efficacy of treatment but the oldest, that of atten-
pounds such as hydrocortisone (cortisol). tion to growth rate and bone age, often are the final
Because an adrenal, testicular, or ovarian tumor measures of adequacy of treatment; if the dose of
may cause virilization in a child and because glucocorticoid is too low, the growth rate is exces-
some metabolites are common to the adrenal sive because of the resulting elevated androgens,
gland and the gonads, the suppression of the whereas too much glucocorticoid quickly sup-
offending androgens by glucocorticoid therapy is presses growth rate and may cause excessive
by itself a diagnostic test for the presence of an weight gain. Bone-age advancement is usually
enzyme defect rather than a neoplasm. While determined yearly; an increase of 1 year of bone
there is good evidence that once per day dexa- age for each year of chronologic age is ideal.
methasone treatment is compatible with normal Serum 17-hydroxyprogesterone measured at a set
172 8 Disorders of Sexual Differentiation

time in the day after a dose of glucocorticoid (usu- At times of stress, glucocorticoid therapy must
ally 2 h after the morning dose in one scheme or at be increased two- to threefold. Usually this will be
8:00 a.m. before the morning dose in another necessary when an infectious illness causes a
scheme) and testosterone (measured in sensitive fever higher than 38.5 °C or 101.3 °F. Current
HPLC MS/MS assays) and androstenedione con- guidelines do not recommend stress therapy for
centrations are frequently used to monitor therapy; minor illnesses such as URIs, athletic activities, or
a 17-hydroxyprogesterone concentration value emotional stress. If the patient is vomiting and
will remain above normal in most cases, but should cannot retain oral medications, the patient should
be well below the untreated state, and age- and be evaluated at the office or emergency room, but
sex-appropriate testosterone or androstenedione on the way if the child appears in extremis, hydro-
concentration are the goals. Salivary steroid deter- cortisone-sodium succinate (Solucortef) can be
minations have been used successfully to study given, intramuscularly or, in case of emergencies
some patients but are not presently in general use. causing shock and poor perfusion, intravenously.
Mineralocorticoid therapy is administered to A dose of 25–50 mg of hydrocortisone-sodium
salt-losing patients in the form of succinate for a child younger than 5 years and a
9-fluorohydrocortisone (Florinef) at 0.05– dose of 50–100 mg for an older child will be an
0.15 mg/day orally. Exogenous mineralocorti- appropriate initial dose for most emergencies. Too
coids cause the retention of sodium and excretion much glucocorticoid in an emergency is not going
of potassium at the distal tubule level of the kid- to cause a problem, whereas inadequate dosage
ney to remedy the lack of endogenous mineralo- may be ineffectual in reversing the effects of adre-
corticoid. Extra salt also may be administered at nal insufficiency and shock.
a dose of approximately 1–2 g/day of sodium Patients with 21-hydroxylase deficiency require
chloride divided into several feedings, which special preparation for surgery to avoid precipitat-
equals 17–34 mEq of Na, since breast milk and ing an acute adrenal crisis. A dose of hydrocorti-
formula have relatively low sodium content; the sone-sodium succinate of 25–100 mg (depending
parents may be given a few test tubes marked at on patient size) is given at the time of induction of
the height 1–2 g of table salt would reach, so that anesthesia, and an infusion of the glucocorticoid is
they may measure the correct amount for the maintained during the procedure, or boluses are
child. Alternatively, a salt tablet can be newly given every 4–6 h to ensure a daily dose in excess
mixed with water or formula and administered in of 45 mg/m2, more than triple the secretory rate of
appropriate milliequivalent dosage. Blood pres- the adrenal glands. This high-dose glucocorticoid
sure can rise high enough to cause hypertensive therapy is maintained during surgery and in case
encephalopathy if too much salt or mineralocorti- of complications, but in most cases, the dose can
coid is given, whereas too little will precipitate be quickly tapered over the several days postoper-
hyponatremia and hyperkalemia and possibly atively. Because of the risks of hyponatremia and
hypotension. Inadequate mineralocorticoid also hypoglycemia, normal saline is infused during the
can be considered a stress and cause elevation of operation and as needed thereafter in a preparation
ACTH and therefore cause increasing androgen with dextrose in 5–10 % concentrations as no
concentrations. In such a situation, where the intravenous florinef is available.
body interprets salt loss as stress, misinterpreta- Patients with 21-hydroxylase deficiency pur-
tion of the androgen levels may suggest the need sue activities in most ways as any other child, but
for additional glucocorticoid, which would lead because they may develop complications of
to growth suppression; if appropriate mineralo- minor illnesses quite easily, they must be observed
corticoid is given instead, the androgens will closely during illnesses. Frequent complications
decrease, and growth will normalize. If control is include hypoglycemia and hyponatremia in salt
poor and bone-age advancement leads to a very losers; any patient with 21-hydroxylase defi-
short predicted adult height, growth hormone has ciency seen with acute illness must be considered
been reported to increase adult height but this is to have hypoglycemia and hyponatremia until
not an FDA approved use of GH. proven otherwise, and initial fluid management
Disorders of Sexual Differentiation 173

must include dextrose and salt. We strongly sug- the child as an adult and informing the family and
gest that patients with 21-hydroxylase deficiency teenager about this years before the transition is
wear an identification bracelet or necklace quite important. Treatment of congenital adrenal
(MedAlert or equivalent) and that they or their hyperplasia of the severe form must continue
parents carry an identifying letter of emergency through life.
procedures from their doctor in case they require
emergency treatment from an institution not Nonclassical CAH Treatment
familiar with this diagnosis. Unfortunately, the Nonclassical CAH is not a condition with DSD
parents must be very assertive as we know of and is discussed in Chap. 9. Treatment of the
informed parents trying to inform emergency nonclassical type of CAH may be continued in
room physicians of the treatment the child should women who experience hirsutism and infertility.
receive but they are dismissed out of hand by the Treatment of males may not be necessary in the
emergency room staff. To anticipate emergencies, adult years according to current guidelines.
it is recommended that all children who leave the
parents for any period of time have a bracelet or Fertility in CAH
necklace stating that they have congenital adrenal In males with CAH the fertility rate is reduced
hyperplasia steroid dependent or perhaps better compared with the normal population, the most
“Addison’s disease” which is a term more easily frequent cause being testicular adrenal rest tumors.
recognized than congenital adrenal hyperplasia in Development and growth of these tumors was
an emergency department and may lead to less assumed to be ACTH-dependent, and it was previ-
confusion. The treatment of hypoadrenal shock ously thought that undertreatment plays an impor-
and hyponatremia is detailed in Chap. 14. tant role. It is now noted these adrenal rests may
Surgical correction of ambiguous genitalia is a increase even if treatment is optimal and that
highly specialized area. Exogenous genitalia are surgical intervention may be considered, but there
ranked by Prader scores ranging from 1 (mild) to is presently no clear indication as to whether
5 (severe) virilization, and the scores generally treatment will affect fertility. Fertility outcomes in
track the severity of the enzymatic defect and the females with CAH depends on their presentation;
degree of salt losing. Clitoromegally may be so classic salt-wasting-type CAH leads to the greatest
severe with a score of 4–5 to require surgery, but degree of problems with fertility. There is a rela-
more often parents now wait for the child to “grow tionship to the degree of adrenal suppression and
into” the size of the clitoris. However, a urogenital the age of menarche and regularity of the menstrual
sinus with vagina and bladder with abnormal cycle. Progesterone and 17-OH-progesterone have
anatomy predispose to urinary tract infections, so to be well regulated in addition to adrenal andro-
imaging is needed to determine if repairs are gens to achieve normal ovulatory cycles. In fact it
required to limit this possibility. As stated above is reported that an important reflection of the
advocacy groups have promoted their strong feel- degree of control is reflected in the regularity of
ings about the surgical treatment of children with menstrual cycles. Polycystic ovarian syndrome,
congenital adrenal hyperplasia and other disor- ovarian adrenal rest tumors, as well as the success
ders with ambiguous genitalia. Hence, consulta- of genital surgery and psychological factors that
tion with experienced pediatric endocrinologists occur from the regular clinic visits and genital
or DSD treatment groups is strongly advised. exams the woman has experienced in her child-
Teenagers with congenital adrenal hyperplasia hood exert influence on fertility. While fertility or
(or other chronic diseases) need plans for transi- desire for fertility is an issue for the adult years of
tion to adult care. The team approach that is so a woman with congenital adrenal hyperplasia, the
important in the care for children with congenital roots of infertility are found during the pediatric
adrenal hyperplasia may be more difficult to find years and the treating physician should be aware
in the adult internal medicine setting (discussed of these issues.
in Chap. 11 regarding transition in diabetes care). Genetic counseling is essential in this and
Nonetheless, considering who will take care of other genetic conditions. The person with con-
174 8 Disorders of Sexual Differentiation

genital adrenal hyperplasia who has a partner present with tertiary hypospadias. The enzyme
sharing the for congenital adrenal hyperplasia deficiency causes a decrease of glucocorticoid
has a one in four chance of having an affected and mineralocorticoid production but an increase
child and a one in four chance of having a com- in dehydroepiandrosterone and its sulfate
pletely unaffected child with a two in four chance (DHEA and DHEAS). As dehydroepiandros-
of having a child who is a carrier of the condition. terone is measured as ketosteroid in the urine,
But genetics is more complex than a simple elevated 24-h urinary ketosteroids, an older test
mathematical schema, hence the recommenda- not used now, will be positive in this condition.
tion for genetic counseling. There will be elevated values of androstenedione
and T concentrations. Due to peripheral conver-
11-Hydroxylase Deficiency sion, 17-hydroxyprogesterone rises and may
11-Hydroxylase deficiency (*202010 ADRENAL lead to a positive CAH screen, but the
HYPERPLASIA, CONGENITAL, DUE TO 17-hydroxypregnenolone/17OHP ratio in the
11-β-HYDROXYLASE DEFICIENCY CYP11B1 basal state or after an ACTH stimulation test (a
At 8q21–q22) leads to virilization, salt retention, rise to more than 100 nmol/l or 3,300 ng/dl) is
hypokalemia, and hypertension; the enzymatic diagnostic. Treatment is the same as with salt-
block leads to excessive 11-deoxycortisol (com- losing 21-hydroxylase deficiency, but the presen-
pound S) and deoxycorticosterone (DOC, a tation may be much more catastrophic in the
potent mineralocorticoid) production, and, newborn due to more severe glucocorticoid and
because ACTH is elevated in the absence of mineralocorticoid deficiencies.
adequate cortisol and because no defect occurs
in the formation of androgens, serum androgens Maternal Sex Steroid Ingestion or
as well as precursors such as 17OHP also are Androgen-Producing Conditions
elevated. Glucocorticoid therapy is necessary to Maternal ingestion of progestins of androgen deri-
stop virilization and reduce salt retention and vation between 8 and 12 of gestation may cause
hypertension. Conversely, mineralocorticoid is 46, XX DSD. Diethylstilbestrol (DES) ingestion is
not used in this condition, as this is a disorder no longer recommended for preventing spontane-
with excessive mineralocorticoid production. ous abortion (it was not effective for this use) since
While diagnosis of girls will usually occur in the 1971 due to its effects on Müllerian duct deriva-
neonatal period due to their ambiguous genitalia, tives leading to clear cell adenocarcinoma of the
those with less severe defects and boys will come uterus and vagina in the daughters of treated
to diagnosis later in childhood due to precocious women. In the sons of treated women DES is
male pubertal development and hypertension. linked to undescended testes, development of cysts
The phenotype of this condition is similar to of the epididymis, and inflammation of the testes.
21-hydroxylase deficiency. There is a late-onset Other exposures to exogenous androgens may
form of 11-hydroxylase deficiency. also virilize the female fetus. If mothers have
androgen-producing tumors of adrenal or ovary,
3-Hydroxysteroid Dehydrogenase the androgens will be passed to the fetus; much
Deficiency of the androgen will be aromatized by the pla-
(*201810 ADRENAL HYPERPLASIA II at centa to estrogen but not necessarily all.
1p13.1) causes salt loss and virilization in
genetic females but inadequate virilization in Other Causes of 46, XX DSD
genetic males, because DHEA is a weak andro-
gen and the only androgen made in the affected P450-Oxidoreductase Deficiency
males. This condition in genetic females does There are also other causes of 46, XX DSD
not cause such severe virilization as in 21-hydrox- such as P450-oxidoreductase deficiency
ylase deficiency, and genetic males are less viril- (PORD) which may be found in the Antley–
ized than the normal male phenotype and usually Bixler syndrome (#201750 ANTLEY-BIXLER
Disorders of Sexual Differentiation 175

SYNDROME WITH GENITAL ANOMALIES leading to absence or severe hypoplasia of the


AND DISORDERED STEROIDOGENESIS; upper two-thirds of the vagina and the absence of
ABS1) due to mutations in the cytochrome the uterus in the presence of normal ovarian func-
P450 oxidoreductase gene (POR, 124015.0001) tion but primary amenorrhea. There are associ-
leading to fetal adrenal androgen excess and ated cervicothoracic somite anomalies, unilateral
ambiguous genitalia in both boys and girls and renal agenesis, and conductive deafness in some
the potential for a host of congenital abnormali- cases. Müllerian aplasia and hyperandrogenism
ties in other organ systems. Antley–Bixler syndrome (158330 MULLERIAN APLASIA AND
type 2 has no genital anomalies (#207410 ICD + HYPERANDROGENISM) are to be differentiated
ANTLEY-BIXLER SYNDROME WITHOUT from androgen resistance; in this case the external
GENITAL ANOMALIES OR DISORDERED genitalia will appear to be phenotypically female
STEROIDOGENESIS; ABS2) is caused by in a 46, XY individual, but the internal ducts will
mutation in the fibroblast growth factor receptor reveal the absence of Müllerian duct derivatives
gene, FGFR2, and presents with craniosynostosis, due to the production of Müllerian duct inhibitory
radiohumeral synostosis, midface hypoplasia, factor and the Wolffian ducts will be absent due to
choanal stenosis or atresia, and multiple joint con- lack of local androgen effect. Thus, testes will be
tractures without steroid abnormalities. present internally, and the patient will present as a
phenotypic female with primary amenorrhea.
Aromatase Deficiency and Estrogen
Receptor Deficiency 46, XX Gonadal Dysgenesis
A loss-of-function mutation in the gene coding 46, XX gonadal dysgenesis (#233300 OVARIAN
for aromatase (#613546AROMATASE DYSGENESIS 1; ODG1) has the phenotype of
DEFICIENCY) eliminates the conversion of an immature female with internal Müllerian
androgens of estrogen and leads to virilization in ducts. The individual has streak ovaries, histo-
a pregnant woman and virilization to some degree logically fibrous tissue, and elevated gonadotro-
in the 46, XX offspring. This is an autosomal pin levels. There is no production of estrogen nor
recessive condition. inhibin. The condition may be sporadic or follow
There are two patients reported with the estro- an autosomal recessive pattern with deafness
gen receptor deficiency (#615363 ESTROGEN (*233400 GONADAL DYSGENESIS, XX
RESISTANCE; ESTRR) in an apparent autoso- TYPE, WITH DEAFNESS).
mal recessive pattern. A 46, XY individual with
absent estrogen receptors will look normal at birth
but with the absence of estrogen will not have nor- 46, XY DSD: Disorders Causing
mal bone-age advancement nor epiphyseal fusion Inadequate Virilization of a Genetic
and will grow to a tall stature and have remark- Male (Previously Called Male
ably decreased bone density. The female patient Pseudohermaphroditism)
as well as the male patient had elevated levels of
estrogen but she also had delayed bone-age devel- An inability to produce testosterone, an inability
opment and no sign of pubertal development. to convert testosterone to DHT in the sexual skin,
or a resistance to androgen action all can cause
Mayer–Rokitansky–Küster–Hauser 46, XY DSD. These are the patients in whom the
Syndrome most uncertainty arises over gender of rearing.
Mayer–Rokitansky–Küster–Hauser syndrome Testicular enzyme deficiencies include some dis-
(%277000 MAYER-ROKITANSKY-KUSTER- orders in which the same enzymes are deficient in
HAUSER SYNDROME) is an autosomal domi- the testes as in the adrenal glands. AMH defi-
nant condition in a 46, XX individual in which ciency will occur in 46, XY DSD conditions with
there is absence of Müllerian duct derivatives abnormal Sertoli cell function which occurs
176 8 Disorders of Sexual Differentiation

when the testes are dysgenetic, but when there tions including adolescent or adult adrenal fail-
are only mutations of the enzymes important in ure. Profound Addisonian crisis will occur by
the production of androgens AMH is normal. 5–7 days after birth because of the complete
absence of mineralocorticoids and glucocorti-
46, XY DSD with Enzymatic Deficiencies coids. Replacement steroid treatment is the
Involving the Gonads and the Adrenal same as for salt-losing 21-hydroxylase defi-
Glands ciency, with the exception that glucocorticoid
replacement dosage may be lower due to the
Smith–Lemli–Opitz Syndrome lack of necessity to suppress androgenic sub-
(#270400 SLOS) is caused by a deficiency of stances produced by the adrenal glands of the
7-dehydrocholesterol reductase (*602858 21-hydroxylase-deficient patient. The condition
7-DEHYDROCHOLESTEROL REDUCTASE; is found in an autosomal recessive pattern and is
DHCR7) leading to deficient steroid produc- most common in Korea, Japan, Switzerland, and
tion including androgens and in some cases some Arab populations.
mineralocorticoids. There are multiple dys- Recently, patients were found with 20–22 des-
morphic features often associated with devel- molase deficiency (#613743ADRENAL
opmental delay, but 46, XY DSD is a frequent INSUFFICIENCY, CONGENITAL, WITH
phenotype although some 46, XY patients look 46, XY SEX REVERSAL, PARTIAL OR
so much like a phenotypic female that diagno- COMPLETE) that had a presentation similar to
sis may be long delayed. StAR deficiency with the exception of absence of
lipoid deposition in the adrenal glands.
StAR Deficiency
StAR (steroid acute regulatory protein) assists 3-Hydroxysteroid Dehydrogenase
the entry of cholesterol into the gonadal or adre- Deficiency
nal cell mitochondria for steroid synthesis. As described above, 3-hydroxysteroid dehy-
StAR deficiency (#201710 LIPOID drogenase deficiency (*201810 ADRENAL
CONGENITAL ADRENAL HYPERPLASIA at HYPERPLASIA II at 1p13.1) in 46, XX DSD
8p11.2) eliminates the production of all adrenal causes salt loss (in most) and inadequate virilization
and gonadal steroid production, and large, lipid- in genetic males. The phenotype is micropenis,
laden adrenal glands develop, which may inferi- perineal hypospadias, bifid scrotum, and a blind
orly displace the kidneys (the condition is also vaginal pouch with gynecomastia often developing
called lipoid adrenal hyperplasia). In the com- at puberty in males. Diagnosis is made by elevated
plete form, all 46, XX and 46, XY individuals pregnenolone, 17a-hydroxypregnenolone, DHEA,
will have a normal female phenotype at birth. and DHEA-sulfate (DHEAS) and elevation of the
The Leydig cells in males will be destroyed by 17OHPreg:17OHP ratio.
the lipoid deposition, while the Sertoli cells will
still produce Müllerian duct inhibitory factor 17α-Hydroxylase Deficiency
so that the female genital phenotype will be 17α-Hydroxylase deficiency (*202110
accompanied by absence of Müllerian ducts. ADRENAL HYPERPLASIA, CONGENITAL,
Remarkably the ovaries in females are not stim- DUE TO 17-α-HYDROXYLASE DEFICIENCY
ulated until puberty so that at the age of puberty at 10q24.3) eliminates the 17-hydroxylation of
sufficient estradiol is produced independent of progesterone and pregnenolone and interferes with
StAR to allow feminization and even anovula- the production of both cortisol and sex steroids;
tory cycles. Genetic males however will have no 46, XX and 46, XY patients cannot produce corti-
progression of secondary sexual development at sol or androgens or estrogens but make excessive
puberty and will remain with an infantile female mineralocorticoid in the form of desoxycorticoste-
phenotype. Incomplete or late-onset forms of rone. The defect is in CPY17, which codes for
StAR deficiency have various clinical presenta- both the 17α-hydroxylase and 17,20-lyase
Disorders of Sexual Differentiation 177

enzymes. Phenotype is of an immature female in albeit undeveloped due to low local testosterone,
the complete form, but some incompletely affected and because of the presence of Müllerian duct
males may have ambiguous genitalia. Thus, hyper- inhibitory factor from the fetal testes, Müllerian
tension, hypokalemia, and elevated serum proges- ducts regress. The upper 2/3 of the vagina is
terone, pregnenolone, corticosterone, and DOC absent (since it is a Müllerian derivative), and the
but decreased PRA, 17-hydroxyprogesterone, and testes are internal or “labial”; patients have had
aldosterone in an immature female phenotype are “hernias” repaired that contained the testes with-
the clinical cornerstones of diagnosis. Primary out the correct diagnosis being made before sur-
amenorrhea in a phenotypic female may be the gery. Gynecomastia or some virilization may
presentation if the subject is first diagnosed in ado- occur at the time of puberty.
lescence. 46, XX patients will have an immature
female phenotype at birth and develop hypergo- 46, XY DSD with Disorders
nadotropic hypogonadism at puberty. of Androgen Action

46, XY DSD with Other Enzyme 5α-Reductase Deficiency


Deficiencies Involving the Testes But 5α-Reductase deficiency (defect in SRDA2, the
Not the Adrenal Gland steroid 5α-reductase enzyme gene *264600
PSEUDOVAGINAL PERINEOSCROTAL
17,20-Lyase or Desmolase Deficiency HYPOSPADIAS; PPSH at 2p23) eliminates the
17,20-Lyase or desmolase deficiency (*202110 conversion of testosterone to DHT in the sexual
ADRENAL HYPERPLASIA, CONGENITAL, skin, thereby causing the formation of chordee
DUE TO 17α-HYDROXYLASE DEFICIENCY (ventral binding of the phallus by attached skin);
at 10q24.3) results from a mutation in the CPY17 small phallic structure; hypospadias; bifid scro-
gene, which causes 17α-hydroxylase deficiency tum, usually with undescended testes; and a uro-
and lack of conversion of 17-hydroxyprogesterone genital sinus; this is described as pseudovaginal
to androstenedione and also 17-hydroxypregnen- perineoscrotal hypospadias. The production of
olone to dehydroepiandrosterone and thereby AMH ensures the absence of Müllerian deriva-
limits the production of testosterone. The CPY17 tives, but the presence of testosterone in high
gene codes for the enzyme that catalyzes the concentrations internally supports the develop-
17-hydroxylation of pregnenolone and progester- ment of Wolffian structures. At puberty, possibly
one, so that deficiency of this action leads to because of increasing production of testosterone
46, XY DSD with decreased 17 OHP values. The or to the effects of steroid 5α-RD type 1, encoded
phenotype is infantile female or ambiguous geni- by the SRD5A1 gene, which is active during
talia with micropenis, perineoscrotal hypospa- puberty and normal SRDA type 1 activity located
dias, and cryptorchidism or “labial” or inguinal in the liver and other nonsexual skin, DHT-
testes. Gynecomastia can occur at puberty and dependent effects such as enlargement of the
some virilization may occur at puberty. penis, descent of the testes are possible (within
the limitations of the anatomy), and pigmentation
17β-Hydroxysteroid Dehydrogenase Type 3 of the scrotum occurs, as well as increased mus-
17β-Hydroxysteroid dehydrogenase type 3 (17β cle mass and deepening of the voice. No acne or
HSD3 or 17β-hydroxysteroid oxidoreductase male pattern recession of hair occurs, however, as
deficiency or 17β-ketosteroid reductase defi- they are dihydrotestosterone dependent. Most
ciency) (#264300 17β-HYDROXYSTEROID remarkably, affected patients from the Dominican
DEHYDROGENASE III DEFICIENCY at 9q22) Republic are initially raised as girls, but at the
interferes with the conversion of androstenedione time of puberty and thereafter are considered
to testosterone and estrone to estradiol. The phe- male in social contexts. This phenomenon has
notype is infantile female or some degree of been considered an example of the plasticity of
ambiguous genitalia. Internal ducts are Wolffian, human gender roles, but the children are usually
178 8 Disorders of Sexual Differentiation

known to have the condition in early life (known female, often tall, with amenorrhea and no pubic
as heuvodoces for the descent of testes at or axillary hair. Thus, a phenotypic female being
puberty), so the situation may not be soley due to evaluated for primary amenorrhea will be
an isolated change in gender. The condition is shocked to be told that she has a 46, XY karyo-
also found in Papua New Guinea, the individuals type during her adolescence or young adult years
are known as kwalatmala, and affected subjects when told in an ill-advised manner: The diagnos-
also change gender role at puberty from female to tician must be aware of such an outcome before
male. It is also reported more frequently in Saudi addressing the issue with the patient and temper
Arabia, but patients are found elsewhere as well. the discussion in a sensitive manner.
Most are raised as boys with reasonable adult LH is usually elevated due to resistance to
adjustment to their lives according to the largest negative feedback from testosterone. AMH is
series of follow-up. Diagnosis is established by normal as the defect is not in the gonads.
measuring the testosterone to dihydrotestoster- The incomplete syndrome of androgen insen-
one ratio in the basal state or after hCG stimula- sitivity is due to a less severe deficiency or less
tion. The testosterone to dihydrotestosterone severe defect in the AR. Phenotype may be nor-
ratio should be less than 10:1. mal male (with the only abnormality being infer-
tility), may be an underdeveloped male with
Androgen Insensitivity small but normally formed phallus, or may be
The complete syndrome of androgen insensitiv- ambiguous. Internal female ducts disappear due
ity (previously called testicular feminization; to normal production of AMH, while male ducts
#300068 ANDROGEN INSENSITIVITY also degenerate due to lack of normal androgen
SYNDROME; AIS at Xq11-q12) results from a action. At puberty, some degree of gynecomastia
loss-of-function mutation in the androgen recep- over the usual amount found in puberty in a nor-
tor gene (AR) on the X chromosome in an mal genetic male may be noted, and some pubic
X-linked recessive pattern. Complete androgen and axillary hair development should occur.
insensitivity is not actually seen as ambiguous Serum LH and testosterone concentrations are
genitalia, because in the absence of testosterone above normal. These patients experience some
action, the phenotype is infantile female. This androgen effect in utero, and reports, albeit lim-
condition is found in a prevalence of about ited, indicate 10–15 % may experience gender
1:20,000–60,000. Internal ducts are hypoplastic dysphoria and chose to change gender either
Wolffian ducts. The vaginal pouch is blind female to male or male to female; the majority
because of the absence of the upper two-thirds of do stay in their initially assigned gender how-
the vagina, normally derived from Müllerian ele- ever. Sex assignment requires the most careful
ments, which atrophies because of the normal consideration and discussion with the parents.
testicular production of AMH, and the uterus and
oviducts are likewise absent due to AMH. In the Unresponsiveness to hCG and LH
newborn period there may be an inguinal hernia Unresponsiveness to hCG and LH (*152790
in a phenotypic girl with masses representing the LUTEINIZING HORMONE/CHORIOGO
gonads. At puberty, the unopposed estradiol NADOTROPIN RECEPTOR; LHCGR at 2p21) is
causes normal feminization, but the lack of associated with low testosterone production and
androgen action eliminates the development of ambiguous genitalia, because in the absence of LH/
pubic and axillary hair. The defect is character- hCG receptors, no Leydig cell testosterone produc-
ized by a decrease in the testosterone and DHT tion can occur. Presentation is phenotypic female if
receptor numbers or a postreceptor defect. complete, but partial absence of LH/CGR allows
Patients usually have an unambiguous female some degree of virilization. Diagnosis is accom-
gender role and are raised as females, so decision plished by showing no substantial rise in testoster-
on sex of rearing is not an issue in the newborn one after hCG administration. Sisters of affected
period. Diagnosis is often made in an adolescent patients may be infertile.
Disorders of Sexual Differentiation 179

Persistent Müllerian Duct Syndrome which can occur with endocrine disorders includ-
Mutations limiting or decreasing the production ing growth hormone deficiency, gonadotropin
of Müllerian duct inhibitory factor, AMH, or its deficiency, and other conditions described in this
receptor (#261550 PERSISTENT MULLERIAN chapter and Chap. 9.
DUCT SYNDROME, TYPES I AND II; PMDS) A notable case of penile ablation due to
are associated with bilateral cryptorchidism in complications of circumcision demonstrates
boys or inguinal uterus and fallopian tubes in the virilizing effect of prenatal androgens on
girls and are inherited in an autosomal recessive central nervous system function; the child was
pattern with male sex limitation. sex reversed to a female gender since there was
no penis but after maturation the individual
46, XX Testicular DSD self-reverted back to a male gender identity.
An individual with 46, XX karyotype may Thus, the decision on sex assignment cannot be
develop testicular tissue if the SRY gene is trans- based on anatomy alone but must include the
located to another chromosome even if there is prenatal exposure to androgens the child might
no Y chromosome (#400045 46, XX SEX experience. The situation again relies upon a
REVERSAL 1; SRXX1). Thus, the SRY gene multiple disciplinary team management as
might be found on X chromosome due to recom- described below.
bination or on an autosome. An individual with
two X chromosomes and an SRY gene may
resemble the phenotype of Klinefelter’s syn- Gonadal DSD
drome. Some 46, XX individuals with reportedly
normal male genitalia or soft testes are found Ovotesticular DSD
with mutations the involving the sox 3 gene Rare patients with both testicular and ovarian tis-
(#300833 46, XX SEX REVERSAL 3; SRXX3). sue have ovotesticular DSD (previously called
true hermaphroditism). The tissue is more often
Anorchia or the Testicular Regression combined in ovotestes on at least one side rather
Syndrome than a normal ovary on one side and a normal
In the testicular regression syndrome a 46, XY testes on the other. Because of variable produc-
individual is found to have no gonads and a small tion of AMH (Müllerian duct inhibitory factor)
but normal scrotum with otherwise normal exter- and local and systemic testosterone, the internal
nal genitalia (%273250 ICD + TESTICULAR ducts and external genitalia will be quite variable
REGRESSION SYNDROME; TRS). While this among individuals. Thus, part of a uterus may be
is discussed in Chap. 9, on occasion the individ- present on one side, with some male ducts on the
ual may have ambiguous genitalia. contralateral side.
The patients most commonly have a 46, XX
Penile Agenesis karyotype, with the SRY present on some chro-
Penile agenesis is a rare sporadic condition which mosomes (but there is no Y chromosome). Other
occurs with abnormalities of development of the cases may have the karyotype of 46, XX/46, XY
caudal structures in some cases. Thus, it may be chimeras or 46, XY individuals with a presumed
found with imperforate anus, absent raphe of the 46, XX cell line in the gonads.
scrotum, and renal defects, often severe. Penile Fertility has rarely been described in ovotes-
aplasia may also occur as an isolated defect in the ticular DSD, but the ovarian tissue can rarely pro-
development of the genital tubercle. Complete duce some feminization at puberty. Conversely,
penile aplasia will not occur with endocrine dis- the dysgenetic testicular tissue can undergo
orders causing decreased biosynthesis or biologi- malignant degeneration and should be removed.
cal effect of androgen. Penile aplasia is not to be Because of concerns over the difficulty of plastic
confused with microphallus in which a perfectly reconstruction of the phallus, most patients are
formed but extremely small penis is present raised as girls.
180 8 Disorders of Sexual Differentiation

46, XY Gonadal Dysgenesis A higher rate of hypospadias in Danish new-


46, XY gonadal dysgenesis (#400044 46, XY borns and smaller testes with lower serum inhibin
SEX REVERSAL 1; SRXY) or Swyer syndrome B, compared with Finish infants, suggested that
may result in an immature female phenotype if environmental agents played a role in the differ-
the dysgenesis is complete or ambiguous genita- ence. This and a high frequency of reproductive
lia if functional Leydig cells remain from the problems among adult Danish men, including
dysgenetic testes. Internal duct formation will impaired semen quality, testicular cancer, and
depend on the amount of AMH produced from increased rate of infantile testicular cancer in a
the dysgenetic testes. The etiology may be abnor- pattern that is attributed to environmental disrup-
malities of SRY function, autosomal mutations, tors, are described as the testicular dysgenesis
or duplication of the Dosage Sensitive Sex rever- syndrome (TDS). There may be a genetic compo-
sal (DSS) locus on the X chromosome. Some nent to susceptibility to the TDS based upon
older individuals may appear to be tall females, GWAS analysis indicating a gene X environment
while others may virilize due to incomplete dys- basis for the disorder.
genesis of the testis. In a remarkable case, an
individual with 46, XY gonadal dysgenesis gave
birth to a phenotypic female with SRY +46, XY The Diagnosis and Treatment
gonadal dysgenesis suggesting that mothers of of Ambiguous Genitalia
children with Swyer syndrome should all be
checked for the presence of Y chromosome mate- A provider who cares for newborns will rarely
rial. The condition may be sporadic or follow have to manage a child with ambiguous genitalia
X-linked male limited (*306100 GONADAL but must always be prepared for that situation.
DYSGENESIS, XY FEMALE TYPE; GDXY), This requires an understanding of the psycholog-
autosomal dominant, or autosomal recessive pat- ical dimensions that will be encountered as well
terns. The dysgenetic testes have a 20- to 46-fold as knowledge of the biochemistry that can cause
increased potential for neoplastic formation and this presentation. While this chapter provides a
must be removed. If an affected subject feminizes basic outline of the physiology of sexual devel-
at puberty, the cause might be estrogen formation opment and the more common conditions that
from a gonadoblastoma formed within the dysge- occur, it cannot provide the experience necessary
netic testes and is a sign of concern rather than to successfully bring the family through the situ-
optimism mistakenly thinking that there is func- ation. While diagnosis is outlined below, it can-
tional ovarian tissue. not be overemphasized how important it is to
Classic 45X Turner presents with normal female immediately obtain a consultation with a pediat-
genitalia phenotype and is discussed in Chap. 9. ric endocrinologist even over the phone before
However, mosaic Turner syndrome with 46, XY/45 entering into detailed discussions with the family
XO karyotype or variants on this mosaic karyotype and embarking upon the diagnostic procedures.
may present with ambiguous genitalia due to the The following paragraphs outline the approach
presence of dysgenetic testicular tissue which may but cannot cover every eventuality nor provide
become neoplastic and must be removed. the reader with full guidance on how to navigate
Klinefelter’s syndrome is a disorder of gonadal the subtleties of family discussions with parents
development but does not present with ambigu- who do not yet know whether they are going to
ous genitalia and is discussed in Chap. 9. raise a boy or girl. In the suggested reading sec-
The differentiation of anorchia from cryptor- tion is a paper in which senior pediatric endocri-
chidism is discussed in Chap. 9. nologists provide advice on the approach to the
infant with ambiguous genitalia that will enhance
the reader’s understanding.
Endocrine Disruptors Ambiguous genitalia in a neonate must be
treated with urgency and consideration. No sug-
Endocrine disruptors are also discussed in Chap. 9. gestion of gender must be made to the parents
The Diagnosis and Treatment of Ambiguous Genitalia 181

until a diagnosis or plan of treatment is estab- Steps in the diagnostic process will vary,
lished. It is appropriate to talk about “the baby” depending on whether the baby has palpable
instead of “he” or “she,” to not have the hospital gonads or whether no gonads are palpable (see
print a card having any suggestion of gender on it, Figs. 8.6 and 8.7). Although a list of appropriate
and to speak of “the gonads” rather than testes or steps exists, it must be emphasized that in this
ovaries. Never say that the baby is “partially girl situation, the help of an expert consultant or DSD
and partially boy.” The parents can be told that the committee is necessary. The vagaries of the inter-
baby has an anatomic problem, that the external pretation of the tests could allow an incorrect
genitalia are “unfinished,” and that it is up to the diagnosis. There is a recent increasing reliance
medical providers, by the way of tests, to deter- upon genetic diagnosis in cases of ambiguous
mine the diagnosis baby and to confer with the genitalia, and genomic methods are becoming
parents as to assigning gender. Even if the physi- easier to order, are less expensive, and have faster
cian has a “gut” feeling about the outcome of the turnaround time. Thus, the classic steroid methods
diagnosis, the risks of incorrect assignment of on these figures may be soon superseded in some
diagnosis are great, and it is best to support the conditions. Ratios of the various steroid metabo-
obviously concerned parents without giving them lites provide further assistance in the diagnosis,
false direction. and standard values for these ratios are available

XX Karyotype XY

Present Absent Present

Low Normal Low

High Normal Low Normal High Normal

Combined 17α−ΟΗ Androstenedione


Salt Losing 17OH Pregnenolone deficiency/17,20
lyase deficiency Testosterone
Low
with hypertension
Normal to Low Normal
Isolated
Moerate Elevation 17,20
No Yes Androstenedione
lyase
DHEAS T/DHT deficiency
P450 Aromatase Normal or CPY17
deficiency or moderate
High
elevation
11 Deoxycortisol High High Normal

Response to
High Normal Testosterone
or normal
receptor
3β-HSD2 Deficiency

Abnormal Normal
11β OH deficiency/ 46, XX DSD
P450c11 deficiency Maternal androgens 5α
with hypertension Ovotesticular DSD Reductase
deficiency
21-OH or P450c21 21-OH or P450c21
Deficiency Virilizing Deficiency salt losing
17-oxidoreductase deficiency Partial androgen 46, XY DSD
or 17βHSD-3 deficiency resistance

Fig. 8.6 Diagnostic algorithm for a patient with ambigu- that play a role in diagnosis, so this is only a basic guide.
ous genitalia in which no gonad palpable; this is not ade- Likewise, new genetic tests can assist in diagnosis and
quate for final diagnosis in many cases. There are many should be investigated on the GeneTests website or other
more considerations based upon ratios of steroid moieties genomic information sources
182 8 Disorders of Sexual Differentiation

Karyotype
XY XO/XY XX,XY,
XX XX/XY

Present Absent Absent Present Present


Mullerian Duct
on Imaging

AMH Low Normal Normal Variable Low Low

17OHP Normal Low Normal High

P450 Oxidoreductase
Combined 17α-OH Testosterone deficiency Ovotesticular
Ovotesticular DSD deficiency/17,20 DSD
lyase deficiency 46, XY DSD
with hypertension
Low Normal

T/DHT
17β-HDS3
deficiency High Normal

5α reductase Response to
deficiency testosterone or
Receptor Assay

Normal Low

Partial Androgen
Ovotesticular DSD
resistance
46, XY DSD

Fig. 8.7 Diagnostic algorithm for a patient with ambigu- ratios of steroid moieties that play a role in diagnosis so
ous genitalia in which gonads (possibly inguinal) are pal- this is only a basic guide. Likewise, new genetic tests can
pable; this is not adequate for final diagnosis in many assist in diagnosis and should be investigated on the
cases. There are many more considerations based upon GeneTests website or other genomic information sources

from the several national endocrine laboratories A phenotypic male must be examined for
identified in Chap. 16 to assess the clinician. phallic size (gently stretched length and width
Medical history is important as in all diagno- eliminating the foreskin), chordee or the binding
ses in pediatric endocrinology. Gestational age, of the phallic structure ventrally, the appearance
birth weight, and maternal exposures are of great of hypospadias, underdevelopment of a scrotum,
importance. Special attention is directed to deter- and palpation for its contents. If testes cannot be
mine if the mother has signs of androgen secre- palpated or measured, a careful examination of
tion or had ingestion of virilizing drugs. The the inguinal area must be performed with lubri-
mother must be queried for a history of cant or soap to smoothly bring the examining
unexplained deaths in the family (which would finger down the inguinal area to see if it can
suggest CAH). Determination of consanguinity move a gonad that is high in the canal into the
is important in the search for autosomal recessive scrotum. In a girl the clitoral size is measured in
conditions. Toxin exposures in the environment width and length eliminating any redundant skin
due to the location of the child’s home or employ- of a clitoral hood and inspecting for scrotaliza-
ment of the parents or other family members who tion of the labia (rugation and pigmentation of
might bring toxins into the house, for example, the skin to look like a scrotum), the appearance
agricultural workers might work among pesti- an “uninhabited” scrotum, or presence of palpa-
cides or other agrichemicals. ble masses in the “labial” area. Labial masses
The Diagnosis and Treatment of Ambiguous Genitalia 183

might be testes, ovaries, or even a uterus. An ful with an ultrasonographer who has experience
examination for asymmetry is essential. with newborns; we have seen cases in which a
Imaging studies are the tests with the quickest completely erroneous reading upset the family
results and may be done soon after a DSD is con- and incorrectly directed the clinician’s efforts.
sidered. An ultrasound performed by an Serum androgen metabolite determinations
experienced radiologist will search for uterus, should be ordered, including at least 17-OH-
ovaries, vaginal appearance and oviducts, and progesterone, androstenedione, DHEAS, testos-
other Müllerian structures in a phenotypic female terone, and DHT. In all cases the laboratory must
or one with ambiguous genitalia. It must be have pediatric standards, and the methods must
emphasized that the radiologist must be experi- be sensitive and specific; for T and DHT the
enced as we have received reports of absence of HPLC/tandem mass spectroscopy method in a
ovaries and uterus in a female child with viriliz- laboratory with pediatric standards is essential.
ing 21-hydroxylase CAH who unequivocally had Thus, with AMH and ultrasound indicating ser-
them. Also the presence of gonads and their toli function and testosterone indicating Leydig
architecture and texture may indicate whether cell function, the defect can be better deter-
testicular, ovarian, or ovotesticular tissue is pres- mined: e.g., both are low in testicular dysgenesis
ent. MRI is sometimes indicated to provide infor- and low T and normal AMH are found in Leydig
mation on structures. Contrast studies of the cell dysfunction due to enzymatic defect.
vaginal opening and urinary tract will reveal National laboratories can run panels, e.g., CAH
sinus formation. In an older child, a bone-age panel, for most or all of these appropriate tests
examination as a reflection of androgen effect is for DSD on small samples in neonates and chil-
useful. Cystoscopy may visualize the anatomy, dren, so do not draw the large samples that your
but in some cases laparoscopy is to be performed local laboratory may think are necessary without
for definitive diagnosis and treatment. checking with the sendout lab. The serum tests
Laboratory determinations will be directed will be done as an emergency if one of the few
toward the possible diagnoses considered. national laboratories experienced in pediatric
Screening for 21-hydroxylase deficiency congen- endocrine testing is used as they understand the
ital adrenal hyperplasia is universal at birth in the importance of this process. Serum LH and folli-
USA, but results may take 7 days to return which cle-stimulating hormone (FSH), if markedly
may be too late for safely initiating treatment if elevated to the castrate range, may suggest dys-
the child has salt-losing tendencies. Likewise, a genetic gonads; sensitive pediatric assays are
screening test is inadequate for diagnosis, so if necessary if low values are to be correctly
there is a high suspicion of 21-hydroxylase con- assessed. It is important to note that gonadotro-
genital adrenal hyperplasia, venous sampling for pins may peak spontaneously in the neonatal
confirmatory laboratory tests must be performed. period and the months thereafter (the minipu-
In most cases karyotype analysis should be per- berty of infancy) and confuse the issue. Urinary
formed as an emergency procedure (laboratories collections for 17-ketosteroid and pregnanetriol
will rush the process in most cases). (As noted, are difficult because of the requirement of a 24-h
no reliance should be put on a buccal smear.) We collection and are no longer used.
must emphasize again that the karyotype will not The diagnosis may be clear from the results,
define the diagnosis but will assist in the determi- although an anxious several-day wait for the
nation of the diagnosis. values will ensue, in even the best situations. If
Anti-Müllerian hormone (AMH) will reflect a question remains whether there is a function-
the presence and function of sertoli cell and will ing testis or what is the activity of 5α-reductase,
partially indicate the state of functioning of the an hCG stimulation test should be performed.
testicular tissue: AMH will be decreased or vari- There are several protocols for the hCG test as
able in syndromes with testicular dysgenesis. An noted in Chap. 9. A single dose of 1,500 IU i.m.
ultrasound of the internal structures can be help- hCG is administered in one diagnostic schema,
184 8 Disorders of Sexual Differentiation

and baseline sample and 72-h serum sample for Unfortunately even with new syndromes
levels of LH, FSH, testosterone, inhibin B, and being discovered regularly, not all children will
AMH are measured. If the testosterone result receive a specific diagnosis.
after 72 h is a level 2–20 times higher than base-
line in infants, the result is normal. If the other
androgen metabolites of interest rise according Treatment
to laboratory standards, an enzymatic defect is
eliminated. If the patient is in mid-childhood or The treatment is tempered to the biological
older, one injection of hCG may not be adequate condition and the potential effects of androgens
to stimulate the quiescent Leydig cells. Thus, on the CNS and is best planned with a multidis-
hCG is given three times per week on alternate ciplinary team. General discussions of treatment
days for 2 weeks, and then measurement of for congenital adrenal hyperplasia are noted
serum testosterone (or other appropriate andro- above in the section regarding 21-hydroxylase
gens or precursors) is performed 24 h after the deficiency. For those conditions that will not
last injection of hCG to determine whether tes- allow maturation at the time of puberty treat-
tosterone secretion can be induced over this lon- ment, follow the plan presented for hypergo-
ger period of hCG administration. Serum levels nadotropic hypogonadism in Chap. 9. As
of DHT as well as testosterone should be mea- emphasized above a transition team for the care
sured if a reflection of the activity of 5α-reductase of the individual as the teenage years are reached
is desired. As another effect of the injections, if is extremely important. The wisdom of interna-
undescended testes are present, the testes may tional senior pediatric endocrinologists is pre-
descend into the scrotum if the inguinal ring is sented in the Wilson reference in the Suggested
not definitively too small to allow the passage. Reading section.
If there is a question of testosterone responsive-
ness of the phallus, 3 months of testosterone
administration should be performed (as noted in The Decision of Sex of Rearing
Chap. 9). Blood or buccal smear for gene analy-
sis or sexual skin biopsy for androgen receptors There are many considerations to determine the
or 5α-reductase activity may be sent to a labora- sex of rearing of the child with ambiguous geni-
tory which performs such testing (see talia. The discussion below is general and cannot
GeneTests), but not all mutations will be be used as a guide book for an individual child.
detected; reported positive mutations range for Added to the considerations below are legal con-
28–90 % depending upon the investigator and siderations. Removal of the gonad in a child that
sporadic versus familial cases. occurs before informed consent can be given has
Serum electrolytes and plasma renin activity many implications. Advocacy groups have
should be monitored in conditions being consid- brought suit against treating physicians who per-
ered with salt wasting. As stated above it may formed gonadectomy or other types of recon-
take several days after birth for hyponatremia structive surgery in children with DSD. This field
and hyperkalemia to develop. is constantly evolving, and before any surgical
Genetic testing for karyotype and fluorescent interventions are done, the advice of the DSD
in situ hybridization for Y chromosome material team as described below and a careful consider-
or SRY will help in many cases and is available ation of all ethical and legal issues are essential.
commercially. Genetic testing for specific diag- The decision about sex of rearing in a patient
noses may be possible: The GeneTests website with ambiguous genitalia depends on the diagno-
lists tests and laboratories as well as indications sis and on the wishes and outlook of the family.
for testing. Since new tests are becoming avail- One consideration is to retain fertility, if possible.
able almost weekly, look at that website for For example, in 21-OH deficiency CAH in a
extremely useful information on the possibility 46, XX individual fertility will usually be possi-
of testing and the location of the laboratories. ble with appropriate treatment, but it is important
The Decision of Sex of Rearing 185

to note that there is a decreased interest in child- scarring. If there is a urogenital sinus, the internal
bearing by affected females with 21-OH defi- anatomy will have to be repaired as well to
ciency, so in some cases, the preservation of decrease the risk of urinary tract infections. The
fertility will not be important to an individual vaginal procedures may be carried out in stages,
(although this knowledge may be clear only in with the final steps done at the time of puberty.
retrospect). Another consideration is to ensure Patients with complete androgen insensitivity
that the external genitalia can be made compati- have a good outcome being raised as females, and
ble with the sex of rearing. However, an overrid- there is no consideration of sex reversal in these
ing issue is the effects of androgens on the fetal patients. 46, XY DSD have usually been raised as
brain as the individual grows which may make, infertile females, except for 5α-reductase defi-
for example, a 46, XY individual with incom- ciency, when male gender role is the usual out-
plete androgen resistance who was raised as a come after puberty. Those 46, XY DSD with
female may experience gender dysphoria and severe testicular enzyme defects or dysgenetic
wish a male role even though the child’s anatomy testes will usually have mainly female phenotypes
led to a decision to raise the child as a girl: and can never make adequate testosterone,
Likewise such an individual raised as a boy on whereas those with partial androgen resistance
occasion may wish to decide to become a female. will be unlikely to respond adequately to testoster-
The parents must be able to cope with the deci- one. Reconstruction of the phallus may be practi-
sion reached, but it may be difficult in some cul- cally difficult if there is no response to androgen.
tures that prize a son to raise a child with 46, XY In all cases of 46, XY DSD, the testes will be
DSD as a daughter no matter what the anatomy undescended or partially descended and will be
may be. On the other hand, some cultures look subject to an increased risk of testicular neoplasm
with disfavor on a daughter who is infertile. in a location where they cannot be regularly
Pediatric surgical or pediatric urological pro- examined. If there are dysgenetic testes, the risk
cedures, performed by an experienced surgeon, rises higher. Thus, orchiectomy has been sug-
are required if anatomic modification is consid- gested in these conditions, again after careful
ered for any patient with ambiguous genitalia. In consideration.
46, XX DSD, the decision will usually be to raise In 5α-reductase deficiency, a potential for fer-
the child as a girl to preserve fertility in spite of tility as a male remains, although reports do not
uncertainty over the later desires over child bear- suggest it is very likely; patients have usually
ing. Most of these patients will have an enlarged been raised as males after correction of the chor-
clitoris, usually too much to appear as a normal dee and hypospadias, and the testes are brought
variation, although there will be relative shrink- to at least the lower inguinal canal where they
age with time and suppressive treatment. In many can be regularly evaluated in case neoplastic
cases the decision will be to let the child “grow change were to occur. In 46Y DSD raised as a
into” the clitoris rather than intervene with sur- male, hypospadias repair may be indicated early
gery. Should a decision be made for modification, to allow him to urinate standing.
the procedure of choice is clitoral recession with In the syndrome of complete androgen resis-
preservation of the glans and the nerve supply, in tance (or testicular feminization in the past), the
contrast to more severe procedures including cli- testes will induce feminization at the time of
torectomy as performed in past decades. Clitoral puberty and are often left in until that time; in the
recession, if indicated, may be performed by age absence of androgen effect there is a lower risk of
1 year to avoid the child being noticeably differ- cancer in these patients before puberty. The
ent from peers in the eyes of babysitters or child- increased psychological difficulty of explaining
care workers, since the child may not always be in the need for a gonadectomy to a teenage child will
the presence of the parents after the first months. be encountered if both explanation of her condition
The vagina may require enlargement, and the pos- to the child and the orchiectomy is delayed. Again,
terior fusion opened in some cases with reevalua- careful consideration of ethical and legal consider-
tion at the time of puberty due to subsequent ations are important if that is the decision.
186 8 Disorders of Sexual Differentiation

If the penis of a boy with microphallus is Youth with Gender Dysphoria


responsive to testosterone, the child can usually
be raised as a male if that is the decision. Patients There is accumulating experience in the appro-
with congenital hypopituitarism and microphallus priate treatment of youth with gender dysphoria
will usually be able to achieve fertility with appro- to improve quality of life. This is not considered
priate therapy and are not considered for sex a disorder of sexual differentiation, and few
reversal (see Chap. 9). affected individuals themselves have a
A question under constant consideration is the DSD. While sex of rearing is assigned at birth or
decision of sex of rearing in a child with partial soon thereafter, gender identity takes many more
androgen insensitivity or with an anatomic defect years to develop, and there may be incongruence.
that makes a male gender assignment far more There are various terms in use by various organi-
difficult. In the past, these patients were raised as zations to define an individual who has a gender
girls. However, increasing evidence indicates that identity different than their sex of rearing with
exposure of the prenatal brain to androgens may gender dysphoria being favored. A person with
cause a tendency to a male gender role, making it gender dysphoria is said to have a marked incon-
more controversial to raise such affected children gruence between the assigned gender and the
in a female gender, without regard for the possi- experienced or expressed gender of at least
bility of reconstructive surgery creating an ade- 6-month duration. Sexual orientation or identity
quate penis. Published cases relate how subjects relates to the choice of the gender of a partner and
were assigned to a female gender but self-reas- may be heterosexual, homosexual, or bisexual,
signed to male gender in the prepubertal or the and any of these occur in gender-conforming per-
pubertal period. Again, the recommendation is to sons as well as gender dysphoric individuals.
consult with a pediatric endocrinologist or DSD Teams with experience in counseling and treating
team to assist in deciding on the sex of rearing youth with gender dysphoria are still rare but are
until this situation is better understood. developing at major medical centers across the
world. Guidelines for their care are available
from various international organizations includ-
Multidisciplinary Team ing the Endocrine Society and are presented in
Management the suggested readings.

If at all possible, a child with DSD requiring con-


sideration of gender assignment should be Suggested Readings
referred to an institution with an experienced
DSD management team, usually located at major 1. Rosenthal SM. Approach to the patient: transgender
youth: endocrine considerations. J Clin Endocrinol
academic medical centers. The composition may
Metab. 2014.
vary but ideally this team will have a pediatric 2. Miller WL, Flück CE. Chapter 13 - Adrenal cortex
endocrinologist, pediatric geneticist, pediatric and its disorders. In: Sperling MA, editor. Pediatric
urologist or surgeon, a psychiatrist or psycholo- endocrinology. 4th ed. Philadelphia, PA: Elsevier;
2014. p. 471–532.e471.
gist with experience in this area, and a legal and a
3. Witchel SF, Lee PA. Chapter 5 - Ambiguous genitalia.
bioethics expert to navigate the thorny issues of In: Sperling MA, editor. Pediatric endocrinology. 4th
parental versus infant rights and informed con- ed. Philadelphia, PA: Elsevier; 2014. p. 107–156.
sent. The experience of the medical team cannot e101.
4. Ostrer H. Disorders of sex development (DSDs): an
be overestimated in importance.
update. J Clin Endocrinol Metab. 2014;99(5):1503–9.
Unfortunately results of the management of 5. Vidal I, Gorduza DB, Haraux E, Gay CL, Chatelain P,
the some conditions, particularly 46, XY DSD, Nicolino M, Mure PY, Mouriquand P. Surgical options
cannot be predicted. The social/psychological in disorders of sex development (dsd) with ambiguous
genitalia. Best Pract Res Clin Endocrinol Metab.
outcome depends not just on medical acumen but
2010;24(2):311–24.
the capability of the caregivers and the individu- 6. Lee PA, Houk CP. Key discussions from the Working
al’s response to the condition. Party on Disorders of Sex Development (DSD) evalu-
Suggested Readings 187

ation, Foundation Merieux, Annecy, France, March 16. Evans JA, Erdile LB, Greenberg CR, Chudley
14-17, 2012. Int J Pediatr Endocrinol. 2013;2013(1):12. AE. Agenesis of the penis: patterns of associated mal-
7. Hughes IA, Nihoul-Fekete C, Thomas B, Cohen- formations. Am J Med Genet. 1999;84(1):47–55.
Kettenis PT. Consequences of the ESPE/LWPES 17. Diamond M, Sigmundson HK. Sex reassignment at
guidelines for diagnosis and treatment of disorders of birth. Long-term review and clinical implications.
sex development. Best Pract Res Clin Endocrinol Arch Pediatr Adolesc Med. 1997;151(3):298–304.
Metab. 2007;21(3):351–65. 18. Ahmed SF, Rodie M. Investigation and initial man-
8. Douglas G, Axelrad ME, Brandt ML, Crabtree E, agement of ambiguous genitalia. Best Pract Res Clin
Dietrich JE, French S, Gunn S, Karaviti L, Lopez ME, Endocrinol Metab. 2010;24(2):197–218.
Macias CG, McCullough LB, Suresh D, Sutton 19. Barbaro M, Wedell A, Nordenström A. Disorders of
VR. Consensus in guidelines for evaluation of DSD sex development. Semin Fetal Neonatal Med. 2011;
by the Texas Children’s Hospital Multidisciplinary 16(2):119–27.
Gender Medicine Team. Int J Pediatr Endocrinol. 20. Ogilvy-Stuart AL, Brain CE. Early assessment of
2010;2010:919707. ambiguous genitalia. Arch Dis Child. 2004;89(5):
9. Lee Y, Styne D. Influences on the onset and tempo of 401–7.
puberty in human beings and implications for adoles- 21. Wilson JD, Rivarola MA, Mendonca BB, Warne GL,
cent psychological development. Horm Behav. Josso N, Drop SL, Grumbach MM. Advice on the
2013;64(2):250–61. management of ambiguous genitalia to a young endo-
10. Reisch N, Rottenkolber M, Greifenstein A, Krone N, crinologist from experienced clinicians. Semin
Schmidt H, Reincke M, Schwarz HP, Beuschlein Reprod Med. 2012;30(5):339–50.
F. Testicular adrenal rest tumors develop indepen- 22. Auchus RJ, Chang AY. 46, XX DSD: the masculin-
dently of long-term disease control: a longitudinal ised female. Best Pract Res Clin Endocrinol Metab.
analysis of 50 adult men with congenital adrenal 2010;24(2):219–42.
hyperplasia due to classic 21-hydroxylase deficiency. 23. Achermann J, Hughes I. Sexual differentiation. In:
J Clin Endocrinol Metab. 2013;98(11):E1820–6. Williams RH, Melmed S, editors. Williams textbook
11. Speiser PW, Azziz R, Baskin LS, Ghizzoni L, Hensle of endocrinology. 13th ed. Philadelphia: Elsevier;
TW, Merke DP, Meyer-Bahlburg HF, Miller WL, 2015.
Montori VM, Oberfield SE, Ritzen M, White PC. A 24. Conte F, Grumbach M. Disorders of sex determination
summary of the endocrine society clinical practice and differentiation. In: Gardner DG, Shoback D, edi-
guidelines on congenital adrenal hyperplasia due to tors. Greenspan’s basic & clinical endocrinology, vol.
steroid 21-hydroxylase deficiency. Int J Pediatr 9. New York: The McGraw-Hill Companies; 2011.
Endocrinol. 2010;2010:494173. 25. Auchus RJ. The classic and nonclassic concenital adre-
12. Rivkees SA. Dexamethasone therapy of congenital nal hyperplasias. Endocr Pract. 2015;21(4):383–9. doi:
adrenal hyperplasia and the myth of the “growth 10.4158/EP14474.RA. Epub 2014 Dec 22. PubMed
toxic” glucocorticoid. Int J Pediatr Endocrinol. PMID: 25536973.
2010;2010:569680. 26. Speiser PW, Azziz R, Baskin LS, Ghizzoni L,
13. Auchus RJ, Witchel SF, Leight KR, Aisenberg J, Hensle TW, Merke DP, Meyer-Bahlburg HF, Miller
Azziz R, Bachega TA, Baker LA, Baratz AB, Baskin WL, Montori VM, Oberfield SE, Ritzen M, White
LS, Berenbaum SA, Breault DT, Cerame BI, Conway PC; Endocrine Society. Congenital adrenal hyper-
GS, Eugster EA, Fracassa S, Gearhart JP, Geffner plasia due to steroid 21-hydroxylase deficiency:
ME, Harris KB, Hurwitz RS, Katz AL, Kalro BN, Lee an Endocrine Society clinical practice guideline.
PA, Alger Lin G, Loechner KJ, Marshall I, Merke DP, J Clin Endocrinol Metab. 2010;95(9):4133–60. doi:
Migeon CJ, Miller WL, Nenadovich TL, Oberfield 10.1210/jc.2009-2631.
SE, Pass KA, Poppas DP, Lloyd-Puryear MA, Quigley 27. Nieman LK, Biller BM, Findling JW, Murad MH,
CA, Riepe FG, Rink RC, Rivkees SA, Sandberg DE, Newell-Price J, Savage MO, Tabarin A; Endocrine
Schaeffer TL, Schlussel RN, Schneck FX, Seely EW, Society. Treatment of Cushing’s Syndrome: An
Snyder D, Speiser PW, Therrell BL, Vanryzin C, Endocrine Society Clinical Practice Guideline. J
Vogiatzi MG, Wajnrajch MP, White PC, Zuckerman Clin Endocrinol Metab. 2015;100(8):2807–31. doi:
AE. Guidelines for the development of comprehen- 10.1210/jc.2015-1818.
sive care centers for congenital adrenal hyperplasia: 28. Houk CP, Levitsky LL. Evaluation of the infant with
guidance from the CARES Foundation Initiative. Int J atypical genitalia (disorder of sex development). http://
Pediatr Endocrinol. 2010;2010:275213. www.uptodate.com/contents/evaluation-of-the-infant-
14. De Felici M. Origin, migration, and proliferation of with-atypical-genitalia-disorder-of-sex-development.
human primordial germ cells. Oogenesis 2013:19–37. 29. Houk CP, Levitsky LL. Management of the infant with
15. Miller WL, Witchel SF. Prenatal treatment of con- ambiguous genitalia. http://www.uptodate.com/con-
genital adrenal hyperplasia: risks outweigh benefits. tents/management-of-the-infant-with-ambiguous-
Am J Obstet Gynecol. 2013;208(5):354–9. genitalia.
Disorders of Puberty
9

ducts producing the fat pad, endothelial cells of


Normal Pubertal Development the blood vessels, as well as fibroblasts. Estrogen
stimulates the pubertal development of the breast.
Puberty should not be considered as a single event Prolactin in adequate amounts can stimulate lac-
but rather a continuous process in which the hypo- tation of the breast, while galactorrhea is a patho-
thalamic–pituitary–gonadal axis develops from logic lactation due to abnormal elevation of
the infantile stage to maturity. This is just one part prolactin secretion.
of the continuum of reproductive function that It is important to note that both boys and girls
begins during fetal life and ends in senescence. can achieve fertility before physical maturity is
The age of puberty is, in large degree (50–80 %), reached.
determined by genetic factors, but environmental In addition to these standard stages of puberty
factors can alter this target. The increasing preva- classically related in textbooks of pediatrics, it is
lence of obesity has been one factor postulated to important to note that the appearance of axillary
decrease the age of menarche in recent years. hair, facial hair, comedones, acne, and axillary
odor are other aspects of pubertal development
which should be noted in the patient’s record. In
Physical Development (Fig. 9.1) addition, although rarely stated in print, the change
in appearance of the face is striking so that young
Physical development of puberty can be denoted children with precocious puberty may have faces
in an objective manner by sexual maturation rat- that more clearly resemble a face of a teenager.
ings (SMR) which are commonly called Tanner The appearance of the vaginal mucosa
stages in honor of the British physician who changes during puberty; the vaginal mucosa
developed these standards. It is important to becomes cornified (keratinified) by estrogen
objectively describe the age of puberty in a devel- effect. The pubertal vaginal mucosa is reddish
oping child so that changes can be appropriately (but not bright red as might be found with an
noted in case the process starts too early, pro- infection) and progresses to a duller pinkish
gresses too quickly, or does not start at all. It is color with clear or slightly whitish discharge
best to separate the stages between pubic hair (not as might appear in an infection, however)
development and other aspects of puberty occurring due to estrogen stimulation. Uterine
(Tables 9.1 and 9.2). volume and ovarian size change with pubertal
Breast tissue is evolved from apocrine glands development and sonographic standards are
and consists of epithelial cells forming the ductal available; uterine volume greater than 1.8 mL
network of the gland, adipocytes surrounding the and ovarian volume greater than 1.2 mL occur

© Springer International Publishing Switzerland 2016 189


D.M. Styne, Pediatric Endocrinology, DOI 10.1007/978-3-319-18371-8_9
A Breast Development and Pubic Hair B Genital Development and Pubic Hair
Breast Pubic hair

Stage 1 Stage 1 Stage 1

Stage 2 Stage 2 Stage 2

Stage 3 Stage 3 Stage 3

Stage 4 Stage 4 Stage 4

Stage 5 Stage 5 Stage 5

Fig. 9.1 (a) Breast development. Stage B1, prepubertal, breast. Stage B5, mature breast with projection of papilla
elevation of the papilla only. Stage B2, breast buds visible only. Appearance of pubic and labial hair among girls.
or palpable with enlargement of the areola. Stage B3, fur- Stage PH1, prepubertal, no pubic hair. Stage PH2, sparse
ther enlargement of the breast and areola with no separa- growth of long, straight, or slightly curly minimally pig-
tion of their contours. Stage B4, projection of areola and mented hair, mainly on labia. Stage PH3, considerably
papilla to form a secondary mount over the rest of the darker and coarser hair spreading over mons pubis.
Normal Pubertal Development 191

Table 9.1 Hormone values in sensitive pediatric assays in puberty


Tanner Boys Girls
Testosterone Estradiol DHEAS
Stage LH (IU/L) FSH (IU/L) (ng/dL) DHEAS (g/dL) LH (IU/L) FSH (IU/L) (pg/mL) (g/dL)
I ≤0.52 <3.0 <5 <89 <0.15 0.50–4.50 <16 <46
II <1.76 0.30–4.00 <167 <81 <2.91 0.40–6.50 <65 15–113
III <4.06 0.30–4.00 21–719 22–126 <7.01 0.40–6.50 <142 42–162
IV 0.06–4.77 0.40–7.40 25–912 33–177 0.10–14.7 0.80–8.50 <283 42–241
V 0.06–4.77 0.40–7.40 110–975 110–510 0.10–14.7 0.80–8.50 See below 45–320
Values according to Quest Diagnostics, 2010; other laboratories will return other standards. Methods used are Estradiol,
ultrasensitive, LCMSMS, in cycling females: follicular stage 39–375 pg/mL, midcycle stage 94–762 pg/mL, luteal stage
48–440 pg/mL; LH-pediatric is an immunometric (sandwich) electrochemiluminescent (ECL) ultrasensitive assay. FSH-
pediatric is an immunometric (sandwich) chemiluminescent ultrasensitive ELISA (C-ELISA) assay. Testosterone is a
tandem mass spectrometry assay; estradiol, ultrasensitive, is a tandem mass spectrometry assay, and DHEA-S is a com-
petitive chemiluminescent immunoassay

in central precocious puberty (CPP) or normal their sixth birthday without apparent disease but
puberty. An experienced sonographer can often with excessive weight. However, it is essen-
determine whether the uterus and ovaries are tial that anyone evaluating a girl showing pubertal
manifesting pubertal stages of development, but development before these lower ages makes cer-
one without experience in pediatrics may erro- tain that there is neither neurological or other cen-
neously report worrisome results. tral nervous system problem nor any premature
primary adrenal or ovarian activity (including a
tumor) causing this early development. The aver-
Age at Onset of Puberty age age of onset of puberty in girls in the USA is
approximately 10 years, and the mean age of
The age of menarche is more easily determined menarche in the USA is about 12.9 years for
than any other aspect of pubertal development, Caucasian girls and 12.3 years for African-
and most historical records show that the age of American girls. Late onset of puberty in girls is
menarche decreased over the last several hundred denoted by lack of secondary sexual development
years (secular trend) to reach a relatively stable by 13 years of age. The accepted lower average
point in the mid-twentieth century. Recently, there age of onset of puberty in boys has not yet changed
is evidence of a decrease in age of menarche once from 9 years in the USA in spite of the obesity
again, and this is often attributed to the obesity epidemic, although recent data shows that over-
epidemic in childhood. Other theories would sug- weight boys begin puberty earlier than lean boys
gest that environmental toxins affect the age of but obese boys may be delayed. The upper age of
menarche as well. African-American girls develop normal pubertal development of boys is 14 years
earlier than Caucasian girls with Hispanic girls of age. A recent study of American boys between
being in between. It is presently accepted that 10 and 16 years of age showed that the mean ages
Caucasian girls may enter puberty following their for onset of Tanner 2 genital development for non-
seventh birthday and African-American girls after Hispanic white was 10.14, African American was

Fig. 9.1 (continued) Stage PH4, thick adult-type hair that darkening of scrotal skin. Stage G5, adult genitalia. Stage
does not yet spread to the medial surface of the thighs. Stage P1, preadolescent, no pubic hair. Stage P2, sparse growth of
PH5, hair is adult type and is distributed in the classic inverse slightly pigmented, slightly curved pubic hair, mainly at the
triangle. (b) Genital development and pubic hair growth base of the penis. Stage P3, thicker curlier hair spread later-
among boys. Stage G1, prepubertal. Stage G2, enlargement ally. Stage P4, adult-type hair that does not yet spread to
of testis to >2.5 cm, appearance of scrotal reddening, and medial thighs. Stage P5, adult-type hair spread to medial
increase in rugations. Stage G3, increase in length and to a thighs (Descriptions from Tanner, Growth at Adolescence,
lesser extent breadth of penis, with further growth of testes. 1962, Oxford, Blackwell Scientific Publications). Adapted
Stage G4, further increase in size of penis and testes and from N Engl J Med 2008;358:2366–2377
192 9 Disorders of Puberty

Table 9.2 Causes and classification of delayed puberty Table 9.2 (continued)
Idiopathic (constitutional) delay in growth and puberty Functional gonadotropin deficiency
Hypogonadotropic hypogonadism: delayed puberty Anorexia nervosa
related to gonadotropin deficiency Bulimia
CNS disorders Psychogenic amenorrhea
Tumor Impaired puberty and delayed menarche in
Craniopharyngioma female athletes and ballet dancers (female athlete
Germinoma triad)
Other germ cell tumors Hypergonadotropic hypogonadism: delayed puberty
Hypothalamic and optic glioma related to gonadal failure
Astrocytoma Girls
Pituitary tumor Syndrome of gonadal dysgenesis (Turner
syndrome) and its variants
Other causes
XX and XY gonadal dysgenesis
Langerhans histiocytosis
Familial and sporadic XX gonadal dysgenesis
Granulomatous and postinfectious lesions of and its variants
the CNS
Other forms of primary ovarian failure:
Vascular abnormalities of the CNS Premature menopause
Radiation therapy Radiation therapy
Congenital malformations especially Chemotherapy
associated with craniofacial anomalies
Autoimmune oophoritis
Head trauma
Resistant ovary
CNS surgery
Galactosemia
Hydrocephalus
Glycoprotein syndrome type 1
Isolated gonadotropin deficiency
FSH receptor gene mutators
With hyposmia or anosmia: Kallmann syndrome
LH–hCG resistance
Without anosmia
Noonan or pseudo-Turner syndrome
Congenital adrenal hypoplasia (DAXI mutation)
Polycystic ovarian disease
Isolated LH deficiency
Boys
Isolated FSH deficiency
Syndrome of seminiferous tubular dysgenesis and
Idiopathic and genetic forms of multiple pituitary its variants (Klinefelter syndrome)
hormone deficiencies
Other forms of primary testicular failure
Other endocrine causes of delayed puberty
Chemotherapy
Hypothyroidism
Radiation therapy
Diabetes mellitus (poorly controlled)
Testicular biosynthetic defects
Cushing disease
Sertoli-only syndrome
Hyperprolactinemia
LH resistance
Miscellaneous disorders
Anorchism and cryptorchidism
Chronic systemic disease and malnutrition
CNS central nervous system, LH luteinizing hormone,
Marijuana use
FSH follicle-stimulating hormone, hCG human chorionic
Gaucher disease gonadotropin
Prader–Willi syndrome
Laurence–Moon and Bardet–Biedl syndromes
Sickle cell disease 9.14, and Hispanic boys was 10.04 years, and for
Cystic fibrosis stage 2 pubic hair, 11.47, 10.25, and 11.43 years,
Acquired immunodeficiency syndrome respectively. Mean years for achieving testicular
Chronic gastroenteric disease volumes of >3 mL were 9.95 for white, 9.71 for
Chronic renal disease African American, and 9.63 for Hispanic boys,
Malnutrition and for >4 mL were 11.46, 11.75, and 11.29
(continued) respectively. For girls, the NHANES III US
The Endocrine Changes of Puberty 193

national study showed the mean ages for onset of it is likely that the boy will still have significant
stage 2 breast development were 9.5 years for growth left. Alternatively, if he has advanced
African Americans, 9.8 years for Mexican pubertal development, he probably has relatively
Americans, and 10.4 years for white Americans, little growth left.
whereas the onset of stage 2 pubic hair occurred at
10.4 years for white Americans, 9.4 years for
African Americans, and 10.6 years for Mexican Skeletal Development and Bone
Americans. Secondary sexual development char- Mineral Density
acteristically spans a period of 4 years from begin-
ning to reaching stage 5. While it is an imperfect method, bone-age deter-
mination gives us a general indication of how
much more growth is available and is often
Ages of Pubertal Stages and Duration invoked to predict adult height. As with ultraso-
nography of the uterus and ovaries, experience is
The Pubertal Growth Spurt extremely important in the accurate interpretation
The greatest growth period in a child’s life occurs of bone-age development. See Chap. 5 for more
during fetal development (one cell to 7 lb in 9 discussion on bone-age assessment.
months!), but there are other growth spurts that The fastest accrual of bone density occurs dur-
are apparent after birth. Increased growth is noted ing infancy and puberty. In America, relatively few
during the first few years in the infant–child teenagers get adequate vitamin D or calcium intake
growth spurt between 1½ and 3 years. The sec- and therefore are at risk for decreased bone den-
ond greatest growth velocity after birth is noted sity, leading to osteopenia or even osteoporosis. A
during the pubertal growth spurt. During this current theory holds that it is possible to achieve
period, girls may gain 25 cm and boys 28 cm. normal bone density later in life by increasing cal-
Girls’ pubertal growth spurts occur about 2 years cium intake after these crucial time periods,
earlier than boys and girls gain less stature during assuming that the individual does not have a
the spurt than boys, and this accounts to the aver- genetic tendency toward osteoporosis. However, it
age difference in adult height between men and is quite important to ensure adequate vitamin D
women of about 5 in. in the USA. and calcium intake as a step to guard against
The control of the pubertal growth spurt decreased bone density during childhood and
depends on numerous factors, including estrogen, puberty and this simple step is often overlooked.
growth hormone, insulin-like growth factors, thy-
roid hormone, and adequate nutrition. In both
boys and girls, it is estrogen, either directly The Endocrine Changes of Puberty
secreted as in girls or converted from testosterone
in boys, that is responsible for the increase in Gonadotropin-releasing hormone (GnRH) is
growth hormone and IGF 1 during the pubertal released in pulsatile fashion from the hypothala-
growth spurt and is linked to the increase in height mus into the hypophysiotropic portal system to
velocity. Estrogen is also responsible for the reach the anterior pituitary which leads to pulsatile
advancement of bone age that ultimately leads to release of gonadotropins, LH and FSH, from the
epiphyseal fusion and cessation of growth. pituitary gland. If, however, GnRH is infused in a
The pubertal growth spurt is an early event in constant manner, the pituitary gonadotrophs will
female puberty, and upon attainment of men- cease to respond to it, receptors for GnRH will
arche, relatively limited growth is left for a girl. decrease, and LH and FSH will fall. This phenom-
The pubertal growth spurt, however, is a later enon is used in the treatment of CPP where long-
event in the puberty of boys. Thus, if a boy com- acting GnRH analogues act similar to a constant
plains of short stature but has not yet entered infusion of GnRH and thereby cause cessation of
pubertal development and has no other disorder, the pubertal level of gonadotropin secretion.
194 9 Disorders of Puberty

While GnRH is one key in the control of centers inhibit the unbridled release of GnRH
puberty, another peptide, kisspeptin, plays an and LH and FSH likewise decrease. In the male
important role. Kisspeptin is secreted from fetus, testosterone secretion is stimulated first by
“KNDy” neurons in the arcuate nucleus that placental human chorionic gonadotropin (hCG).
secrete neurokinin B and dynorphin as well. hCG levels fall by week 20 and thereafter fetal
Kisspeptin interacts with its receptor, KISSR pituitary luteinizing hormone secretion stimu-
(previously called GRP-54), to stimulate GnRH lates the fetal testis. Normal penile development
secretion. Mutation of the receptor or absence of requires hCG and a functional testes early in
kisspeptin protein will both eliminate pubertal development, but if there is no pituitary LH avail-
development and reproduction. Alternatively, an able by midgestation, the penis, although perfectly
excess of kisspeptin or a stimulatory mutation of formed, will not enlarge to normal size and a new-
GRP-54 will cause precocious puberty. born boy with gonadotropin deficiency will have a
Pituitary follicle-stimulating hormone stimu- microphallus (or a phallus length more than 2.5
lates the follicles of the ovary to produce estrogen SD below the mean which is less than 2.0 cm in
and the seminiferous tubules of the testes to sup- stretched length (although some sources state less
port sperm development. Luteinizing hormone than 2.5 cm defines microphallus)). A microphal-
has less effect on the female ovary until ovulation lus will also be narrower than a normal penis.
occurs, at which time it stimulates the luteiniza- After birth, when the newborn infant is
tion of the follicle into the corpus luteum. In a boy, released from the inhibitory effects of high con-
luteinizing hormone stimulates the Leydig cells to centrations of maternal estrogen, pulses of pitu-
produce testosterone. The Sertoli cells of the tes- itary LH and FSH increase in amplitude and
tes and the follicles of the ovaries also produce the frequency, leading to pulses of steroid secretion
protein hormone inhibin which, in addition to sex from the gonads. Infantile testosterone concen-
steroids, exerts a feedback inhibitory effect on LH trations can peak as high as 150 ng/dL during this
and FSH secretion. The inhibin molecule has a “mini-puberty of infancy,” a clearly mid-pubertal
beta subunit termed activin; activin stimulates the level in normal boys. Girls likewise have elevated
synthesis and release of FSH from the pituitary peak levels of estradiol that can bring about
gland. Follistatin, produced in the pituitary gland, breast development soon after birth. Thus, gonad-
inhibits the activin effect by binding to the activin otropin values reach pubertal levels during the
receptor. The testes likewise produce anti-Mülle- first months and years after birth, and sex steroid
rian hormone (AMH, sometimes called Müllerian secretion in both boys and girls can episodically
duct inhibitory factor or MIF; see sexual differen- reach values as high as those found during
tiation Chap. 8). Measurement of the protein prod- puberty; this makes the diagnosis of precocious
ucts of the gonads is useful to determine the puberty in infancy all the more difficult.
presence or absence of gonads and may assist in The juvenile pause (childhood phase of
the differentiation of constitutional delay in decreased endocrine activity after the mini-
puberty (CDP) from idiopathic hypogonado- puberty of infancy) that follows infancy is char-
tropic hypogonadism (IHH). Gonadotropin acterized by low activity of the hypothalamic
secretion is very high in primary gonadal failure GnRH secretion and low levels of LH and FSH
due to the absence of inhibin as well as the secretion. Thus childhood gonadotropin secre-
absence of sex steroids so that there is diminished tion is also pulsatile, but the amplitude is quite
negative feedback inhibition occurring. low. If the child has no functional gonads, such as
Hypothalamic GnRH begins to stimulate pitu- is found in Turner syndrome, gonadotropin con-
itary LH and FSH release in the fetus. By midg- centrations rise higher than normal, although
estation when the hypothalamic pituitary portal they do not rise as high as the levels found later in
system is functional and hypothalamic GnRH Turner syndrome during the pubertal age range.
can reach the pituitary gland, LH and FSH con- This indicates that intrinsic CNS inhibition is
centrations are extremely high. As fetal develop- active during the childhood juvenile pause with
ment progresses, higher central nervous system feedback inhibition by sex steroids and inhibin
The Endocrine Changes of Puberty 195

active at a high level; the strength of the feedback lating testosterone secretion. Likewise elevated
inhibition decreases just before and continues gonadotropin levels will stimulate infantile ova-
during the pubertal period allowing gonadotro- ries for a period of months after birth. At the age
pins and sex steroids to rise to pubertal levels. of onset of puberty decreased inhibition of GnRH
The control of the intrinsic CNS inhibitory secretion by higher central nervous system
process suppressing GnRH secretion appears (decreased inhibition by GABA and MRKN3 is a
mediated by various CNS peptides including major mechanism) and decreased sensitivity to
gamma-amino butyric acid (GABA), an inhibitory negative feedback inhibition by sex steroids and
neuropeptide. The recent discovery of MKRN3 a inhibin allow increased GnRH secretion into the
paternally expressed maternally imprinted gene, pituitary portal circulation. Thus, to a degree
as a putative “brake” on the endocrine control of puberty might conceptually be considered to be a
puberty provides another clue as to the control condition which is held back from a younger
of the onset of puberty as inactivating mutations of child until the higher level inhibition is with-
MKRN3 lead to familial CPP. drawn. Congenital defects of the hypothalamus,
CNS surgery, or injury to the hypothalamus can
release this inhibition and bring about CPP. After
Gonadarche (Fig. 9.2a) the inhibition of puberty during the juvenile
pause, at the time of peripubertal period or the
Gonadarche denotes the reawakening of the onset of puberty there is an increase in stimula-
gonad at the time of pubertal development after tory peptides such as kisspeptin and glutamate
the juvenile pause due to stimulation by increas- which increase GnRH secretion in turn.
ing amplitude and frequency of GnRH pulses, Plasma gonadotropins first rise at night in a
leading to increased pulsatile pituitary gonado- diurnal rhythm in the peripubertal period, the
tropin secretion. Gonadotropin secretion is prom- period just before physical development begins,
inent in the fetus once hypothalamic pituitary and during later periods of pubertal development,
portal system develops by midgestation. they rise during the day as well until there is no
Unrestrained gonadotropin secretion leads to longer any diurnal variation; adults follow this
stimulation of fetal testosterone secretion in the pattern of relatively equal secretion during day
male. During gestation the fetal adrenal zone of and night. Clinically it would be better to measure
the adrenal gland produces DHEA which is gonadotropin secretion in the middle of the night
metabolized by the placenta to produce estriol to determine if puberty has begun but practicality
and other estrogens which are present in high necessitates measurement during clinic hours.
concentrations. The placenta likewise produces The episodic increased amplitudes of LH and
large amounts of progestin; this would suppress FSH secretion are followed within a few hours by
maternal gonadotropins inhibiting ovulation increased sex steroid secretion which leads over
during pregnancy. Progestin also supports the time to the physical changes of puberty described
continuation of the pregnancy by supporting above.
the endometrium and suppressing myometrial Due to the increase in gonadotropin levels the
contractility during gestation. As gestation pro- gonads secrete their respective sex steroids. The
gresses, inhibitory substances including gamma- ovary secretes estrogens and the testis secretes
aminobutyric acid (GABA) and possibly the testosterone either of which brings about the
products of the gene MKRN3 suppress central physical changes of pubertal development. In the
nervous system stimulation of gonadotropin female estrogen secretion leads to breast devel-
secretion. Soon after birth and the removal of the opment while adrenarche (described below) with
suppressive action of maternal estrogen, gonado- rising adrenal androgens lead to axillary and pubic
tropins peak once again and remain elevated for hair development as well as acne. In the male tes-
months after birth in both boys and girls. The tes enlarge as a reflection of increased seminifer-
male infant will experience the mini-puberty of ous tubular development and to a lesser extent
infancy due to this gonadotropin secretion stimu- Leydig cell development. The testes produce
196 9 Disorders of Puberty

a Third ventricle Hypothalamus

Neuroendocrine
cell nuclei

GnRH secreting cells

Superior
hypophyseal Stalk
artery

Long portal
vessels Inferior
hypophyseal
artery
LH and FSH

Secreting cells

Anterior Posterior
pituitary pituitary

Hormone Short portal


secretion vessel

+ LH and FSH

Testosterone Estrogen
and inhibin and inhibin

Testes Ovary

Fig. 9.2 (a) The normal physiologic state of gonadotro- the hypothalamus does not produce gonadotropin-releasing
pin-releasing hormone episodically stimulating pituitary hormone (top “X” indicating tertiary hypogonadotropic
gonadotropins to stimulate the gonads to release sex ste- hypogonadism) or the pituitary gland does not release
roids which exert negative feedback on the pituitary and gonadotropins (lower “X” indicating secondary hypogo-
hypothalamus to reach an equilibrium. The negative feed- nadotropic hypogonadism) so that the gonads do not
back equilibrium will be set at a higher sensitivity in secrete their steroid hormones. (c) Hypergonadotropic
childhood and a lower sensitivity during puberty. The hypogonadism in which the gonads do not produce sex
gonads also make a protein hormone, inhibin, which steroids nor inhibin leading to extremely elevated levels
exerts negative feedback inhibition but is not shown in the of LH and FSH due to the lack of feedback inhibition
diagram. (b) Hypogonadotropic hypogonadism in which

mainly testosterone but produces a small amount by estrogens and decreased by androgens.
of dihydrotestosterone and DHEA. Testosterone Besides androgens, the testis also produces estro-
circulates with albumin (38 % roughly) or sex gens. Estrogen receptors are present in the male
hormone-binding globulin (60 %) leaving 2 % as epididymis and spermatic ducts and apparently
free testosterone which can diffuse into the tis- play a prominent role in maintaining fertility. The
sues. Sex hormone-binding globulin is increased majority of estrogen in the circulation in males
The Endocrine Changes of Puberty 197

b Third ventricle Hypothalamus

Neuroendocrine
cell nuclei

GnRH secreting cells

Superior
hypophyseal Stalk
artery

Long portal
vessels Inferior
hypophyseal
artery
LH and FSH

Secreting cells

Anterior Posterior
pituitary pituitary

Hormone Short portal


secretion vessel

LH and FSH

Testosterone Estrogen
and inhibin and inhibin

Fig. 9.2 (continued)

however is produced by aromatase acting on cir- single-androgen receptor to combine with


culating testosterone. Testosterone and dihy- another receptor molecule; thus, androgen and its
drotestosterone binds to the androgen receptor receptor translocate into the nucleus, binding to
which is coded on the X chromosome. DNA and ultimately transcription and protein
Dihydrotestosterone binds more avidly to the formation of androgen action occur.
receptors than testosterone which accounts for its Antiandrogens compete with androgen bind-
increased potency. This receptor like other ste- ing to the androgen receptor. Cyproterone acetate
roid receptors is a member of the steroid–thyroid is a steroid molecule that acts as an antiandrogen
hormone nuclear superfamily, found in the cyto- while flutamide, bicalutamide, and nilutamide
plasm, and associated with several heat-shock are nonsteroidal antiandrogen antagonists. Anti-
proteins and immunophilins. When testosterone antigens are used in girls with hirsutism or in
or dihydrotestosterone binds to the receptor, it conditions such as premature Leydig cell and
dissociates heat-shock proteins from the complex germ cell maturation in boys to decrease andro-
of the androgen dimerized receptors allowing a gen effects if the source of androgens cannot be
198 9 Disorders of Puberty

c Third ventricle Hypothalamus

Neuroendocrine
cell nuclei

GnRH secreting cells

Superior
hypophyseal Stalk
artery

Long portal
vessels Inferior
hypophyseal
artery
LH and FSH

Secreting cells

Anterior Posterior
pituitary pituitary

Hormone Short portal


secretion vessel

LH and FSH

Testosterone Estrogen
and inhibin and inhibin

Fig. 9.2 (continued)

removed. Antiandrogens have been used in con- hormone-binding globulin (38 %) and albumin
junction with glucocorticoids in the treatment of (60 %) with about 2 % circulating his free estra-
virilizing CAH. Antiandrogens are teratogenic. diol. Estradiol is either directly conjugated or
Estrogen is produced by the granulosa cells of converted into estrogens and then conjugated
the ovarian follicles while progesterone and before excretion in the urine.
androgens are produced by the theca cells which Estrogens stimulate mammary gland develop-
lack aromatase that could convert such androgen ment in puberty. Estradiol also has effects on
precursors into estrogens. The ovaries secrete fluid balance as it lowers the point at which argi-
one-third of woman’s testosterone production, nine vasopressin is secreted and at which thirst
the other two-thirds being produced from periph- arises and increases sodium resorption in the kid-
eral conversion from precursors. About 80 % of ney, thereby increasing plasma volume.
circulating estradiol comes from the ovary with Selective estrogen receptor modulators
the rest from peripheral effects of the aromatase (SERMs) block estrogen effects to a different
enzyme. Circulating estradiol is bound to sex degree in different tissues. Because estrogen
The Endocrine Changes of Puberty 199

advances bone age and causes epiphyseal fusion, androstenedione and testosterone from the ova-
various approaches to delay the effects of estro- ries and the adrenal glands. Androstenedione can
gen on these processes are utilized in clinical be converted to testosterone by many cells in the
studies. Thus tamoxifen, ormeloxifene, and ral- body. In the theca cells androstenedione produc-
oxifene act as antagonists on bone (also act as tion is under the control of LH; androstenedione
antagonists at breasts and uterus). Clomiphene is converted to estrogen in neighboring granu-
acts as an estrogen antagonist on the hypothala- losa cells under the control of FSH. But in poly-
mus but also exerts some estrogen effects; it can cystic ovarian syndrome, there is predominant
be used to stimulate secretion of LH and FSH by secretion of androstenedione from the ovaries.
blocking estrogen negative feedback inhibition 17-Hydroxyprogesterone, a precursor of andro-
on the hypothalamic–pituitary axis. Aromatase gens, is produced by theta cells of the ovary during
inhibitors stop the production of estrogen from the luteal phase and the Leydig cells of the testes.
androgens and have been invoked to block the The majority of dihydrotestosterone is pro-
effect of estrogen on bone maturation in order to duced by peripheral conversion by 5-alpha-hydrox-
increase adult height. ylase of testosterone usually in localized tissue.
Progesterone is the product of the theca cells Testosterone production in the testes leads to
of the luteinizing follicle during the luteal enlargement of the phallus. Conversion of testos-
phase of the menstrual period. It is also a mol- terone to the highly potent androgen dihydrotestos-
ecule important in the metabolic production of terone usually locally leads to the production of
other steroid molecules including cortisol, min- pubic and axillary hair and male pattern baldness
eralocorticoids, and androgens. Progesterone is as well as prostate growth. Testosterone is metabo-
also a product of the testes, is a precursor in the lized by oxidation, reduction, hydroxylation, and
biosynthesis of testosterone, and also has finally conjugation but may be excreted in the urine
effects on spermatogenesis and Sertoli cell as other steroid hormones are.
function. Severely decreased weight is incompatible
Progesterone acts on its receptor (PR, also with menarche or even pubertal development due
known as NR3C3 or nuclear receptor subfamily 3, to decreased secretion of GnRH and therefore of
group C, member 3), in a manner similar to other LH and FSH. There has been considerable specu-
steroid hormones such as estrogen described above. lation as what factor may cause low weight to
Progesterone increases plasma volume by lead to decreased gonadotropin secretion. Leptin,
increasing protein retention in the intravascular the hormone released by adipose tissue (see
space. Progesterone does not appear to exert the Chap. 13), appears to be a factor which is neces-
same effect on lowering the threshold for thirst sary, but not sufficient, for pubertal development.
and vasopressin secretion as estrogen. The combi- In the absence of leptin effects, in the rare indi-
nation of some progesterone forms and estrogens viduals who are deficient in leptin production
in an oral contraceptive limits fluid retention. (#614962 LEPTIN DEFICIENCY; LEPD) or in
Progestin androgenic activity varies with prep- leptin receptors (#614963 LEPTIN RECEPTOR
aration. Low androgen is found in norgestimate, DEFICIENCY), puberty does not occur demon-
desogestrel, norethindrone 0.4–0.5 mg, and strating the importance of leptin in the pubertal
drospirenone; moderate androgen is found in levo- process. Both of these are autosomal recessive
norgestrel triphasic, norethindrone acetate 1.0 mg, conditions. When leptin is administered in leptin
norethindrone 1.0 mg, and ethynodiol diacetate deficiency, puberty will progress if the subject is
1.0 mg; and high androgen is found in norgestrel of pubertal age or bone age is appropriate for
0.3 mg, norethindrone acetate 1.5–2.5 mg, and pubertal development: Leptin will not cause chil-
levonorgestrel 0.15 mg. dren to advance into puberty in isolation.
Androstenedione is a precursor of androgens Stress suppresses gonadotropin secretion.
in the adrenal glands, testes, and ovaries. In nor- This may be one mechanism of hypothalamic
mal women there is an equal production of amenorrhea.
200 9 Disorders of Puberty

Spermarche because of ACTH deficiency or alternatively due


to enzymatic defects blocking androgen produc-
Spermarche is the first appearance of sperm, tion (see Chap. 8). In general, adrenal androgens
noted in first morning urine specimen in clinical are not used for therapy in pediatric or adolescent
studies, and occurs at a mean chronologic age of patients, but DHEA is available for treatment
13.3–16 years depending upon the study, and at a should there be a complaint about inadequate
mean pubic hair stage 2–3. However, spermarche sexual hair development.
may be found in normal boys with bilateral tes- Adrenarche characteristically is described as
ticular volumes of only 3 mL and no physical an event that occurs at 6 and 8 years of age;
signs of puberty. increasing secretion of adrenal androgens actu-
ally starts around 3 years of age and progresses
from there. Premature adrenarche is usually
Adrenarche characterized as onset of pubic hair before 6–8
years of age and exaggerated adrenarche is used
Adrenarche denotes the increase in adrenal as the term if androgen values are higher than
androgens that occur prior to the appearance of characteristically found in the more common
pubic hair which is called pubarche; adrenal form of premature adrenarche.
androgen values rise before gonadotropin secre- There is usually coordination between gonad-
tion increases. Adrenarche only occurs in human arche and adrenarche, but the control for each is
beings and our closest primate relatives. There is distinct and either can occur first at the time of
an increase in the size of the zona reticularis puberty. If the rise in adrenal androgens occurs
associated with the increase in dehydroepian- earlier than in the normal progression, premature
drosterone, dehydroepiandrosterone sulfate, and adrenarche and subsequent premature pubarche
androstenedione secretion, the major androgens occur. The age of classic premature adrenarche
of the adrenal. These are weak androgens that are described in the medical literature may overlap
converted to more potent androgens such as an age that is now considered within the normal
testosterone which itself can be converted by limits of the onset of pubertal development, so
5-alpha-hydroxylase to dihydrotestosterone. confusion may occur as to whether an individual
Adrenal androgens also can be converted to has normal or premature adrenarche.
estrogens of various potencies by the enzyme
aromatase. Adrenal androgens circulate primar-
ily bound to albumin with a smaller contribution Pediatric Sex Steroid
from sex hormone-binding globulin. In addition and Gonadotropin\Assays
to the appearance of pubic hair, there is a change
in complexion with the onset of comedones or Assays aimed toward the adult population gener-
acneiform lesions and increased axillary hair; ally are calibrated to indicate if LH and FSH are
adrenal androgens may rise early in the condition very high, as found in postmenopausal women,
premature adrenarche; often growth rate increases and whether testosterone is as high as in a normal
slightly, although not as much as found in preco- adult male or low as found in testicular failure.
cious puberty, and bone age may be slightly Specialized pediatric assays are necessary if one
advanced over the normal 2 standard deviation is to know the true values of sex steroids and
variation. Adult height is characteristically gonadotropins in childhood or during pubertal
reported as normal even with some advancement development. Thus, testosterone should be mea-
in bone age. Alternatively, the absence of adrenal sured by HPLC tandem mass spectroscopy which
androgens leads to a diminished development of usually is carried out in a national lab (since the
pubic and axillary hair, but the androgen produc- equipment is extremely expensive) which has
tion of the testes and the ovaries lessens that pediatric standards. This assay is more expensive
affect. Adrenal androgen deficiency may occur than the standard adult testosterone assay but is
The Endocrine Changes of Puberty 201

the commercial assay that is valid in childhood diagnosis of the impending onset of puberty in
and in women as well. If a sample is subject to constitutional delay in puberty.
extraction and chromatography and then mea- More than 97 % of sex steroids are non-
sured in a radioimmunoassay, valid values for covalently bound to sex hormone-binding globulin
evaluation of puberty will result, but this modi- (SHBG), sometimes denoted testosterone-binding
fied radioimmunoassay is not available in most globulin (TEBG). Estrogen stimulates a rise in
laboratories without personally requesting the SHBG, which has the effect of increasing the
special steps to be carried out. It is possible to protein-binding of testosterone so that a given
measure free testosterone which is not bound to level of testosterone has less biological effect in
sex hormone-binding globulin in bioavailable girls with higher SHBG. Androgens in boys,
testosterone assay; bioavailable or bioactive tes- however, decrease SHBG, thereby allowing more
tosterone assays are useful in the evaluation of unbound and therefore more active testosterone to
virilizing disorders in girls and women. Estradiol circulate in a manner which increases androgen
is best measured in the HPLC tandem mass spec- effect in a boy as pubertal development progresses.
troscopy system as well. LH and FSH should be The girl who has increased androgen levels due to
measured in a highly sensitive immunometric polycystic ovarian syndrome will have decreased
assay and interpreted with pediatric standards, SHBG which likewise will increase the effects of
again characteristically requiring a national send- her androgen levels.
out laboratory. There is virtually no useful infor- Inhibin B rises as Sertoli cells mature during
mation if the standard LH and FSH assay is puberty. Conversely AMH decreases in concert
carried out in many disorders of puberty since with the rise in inhibin as the Sertoli cells mature.
they are far too insensitive to accurately deter- Both can yield important information in the
mine childhood values; they could be used in evaluation of puberty or disorders of sexual
gonadal failure such as in a pubertal aged girl differentiation.
with Turner syndrome or a late pubertal boy with
Klinefelter syndrome, but not in the normal child
or one with a more subtle problem. Ovulation and Menarche
As pubertal development begins, a larger store
of gonadotropins develops in the pituitary gonad- The last stage in hypothalamic–pituitary devel-
otrope cells. Stimulation of the pituitary gonado- opment is the onset of positive feedback, leading
trophs with GnRH, or more recently with GnRH to ovulation and menarche. While in prepuberty a
agonists which are more readily available, allows small amount of estrogen will suppress LH and
determination of maximal gonadotropin secre- FSH secretion, after midpuberty, estrogen in the
tion, an important diagnostic criterion for the appropriate amount at the appropriate time in the
onset of puberty. However, the highly sensitive menstrual cycle can stimulate gonadotropin
pediatric LH and FSH assays can be used on sin- release. Thus, as puberty progresses increasing
gle, unstimulated blood samples to determine the estrogen secretion from the ovary occurs as feed-
onset of puberty, sometime eliminating the need back inhibition of estrogen on gonadotropin
for a GnRH stimulation study to determine the secretion decreases. Primordial follicles are the
presence or absence of pubertal gonadotropin fundamental reproductive units that comprise the
values. Pediatric ultrasensitive gonadotropin and pool of resting oocytes. Primary follicle develop-
sex steroid determinations are capable of demon- ment is the first stage of follicular growth.
strating increasing values before physical puber- Primary follicles differ from primordial follicles
tal development is detected. in several ways as the oocyte begins to grow.
In boys, an 8:00 a.m. serum testosterone value As growth progresses, the zona pellucida is
of 20 ng/dL, or 0.7 nmol/L, has been shown to formed and the secondary follicle develops which
herald the onset of male pubertal development includes attainment of maximal oocyte growth,
within the next 12 months, a finding useful in the proliferation of granulosa cells, and acquisition
202 9 Disorders of Puberty

of theca cells. The tertiary follicle enters the early menstrual bleeding occurs more than every 21
antral phase characterized by the formation of an days or lasts more than 7 days or soaks more than
antrum or cavity in the follicle. The antral fluid one pad or tampon every 1–2 h. Minor irregulari-
contains steroids, proteins, electrolytes, proteo- ties of the menstrual cycle are usually due to
glycans, and an ultrafiltrate that forms from dif- abnormal hypothalamic pituitary ovarian func-
fusion through the basal lamina. Other changes in tion and often resolve while more serious and
this phase include further theca cell differentia- long-lasting complications require evaluation
tion. Subpopulations of thecal interstitial cells and treatment. The high prevalence of anovula-
develop within the theca interna, acquire LH tory periods after menarche may be due to unrec-
receptors, and are capable of steroidogenesis. ognized PCOS in some individuals rather than a
The granulosa cells begin to differentiate into normal developmental phenomenon. It is recom-
distinct cell layers. The dominant follicle develops mended that significant menstrual irregularity be
which is able to increase estrogen secretion evaluated if it persists over 1 year and especially
during the follicular phase or proliferative phase that is associated with other pathologic findings
of the menstrual cycle to exert positive feedback such as galactorrhea or hirsutism. However, some
on the hypothalamus leading to the midcycle LH of the first cycles after menarche may be ovula-
surge that brings about ovulation. Once ovulation tory, and fertility is possible in the first cycle.
occurs, LH causes luteinization of the granulosa Just as in boys, girls are reproductively mature
cells of the follicle leading to a rise in progester- prior to physical maturity; girls may become fer-
one as well as estrogen during the luteal or secre- tile and even pregnant prior to physical or emo-
tory phase. Finally, estrogen falls again along tional maturity.
with progesterone and the menstrual cycle occurs.
During the follicular phase the uterus builds up
by proliferation and differentiation of the endo- Miscellaneous Metabolic Changes
metrium until the functional layer is shed and
menstrual flow occurs. However, even if the mid- The rise of sex steroid concentrations at the
cycle surge of gonadotropins is present, ovula- onset of puberty is either directly or indirectly
tion may not occur during the first menstrual associated with many changes in other labora-
cycles; 85–90 % of menstrual cycles are anovula- tory values. In boys, the hematocrit rises. In
tory in the first year after menarche, about 60 % both boys and girls high-density lipoprotein
in the third year after menarche, and about 25 % (HDL) concentrations rise and alkaline phos-
in the fifth year after menarche. Menstrual irregu- phatase rises during the pubertal growth spurt
larities in normal girls appear to be relatively (often this rise is incorrectly interpreted as evi-
minor as menstrual length variation is 21–45 dence for a prostatic tumor or liver abnormal-
days in 75 % of girls during the first menstrual ity). Serum IGF-I concentrations rise with the
year, but this variation decreases to about 5 % by growth spurt. Prostate-specific antigen (PSA) is
the third menstrual year. measurable after the onset of puberty in boys and
Recently revised definitions of abnormal uter- may provide another biochemical indication of
ine bleeding state that secondary amenorrhea pubertal onset.
occurs when no menstrual cycle appears in a
period of over 90 days after menarche, and oligo-
menorrhea occurs when there are fewer than four Abnormalities of Puberty
periods in the year (average cycle length 90 days)
in the first year after menarche, fewer than six Delayed Puberty
periods in the year (average cycle length 60 days)
in the second year after menarche, and fewer than If a girl does not begin any aspect of pubertal
eight periods per year (average cycle length 45 development by 13 years, or a boy by 14, the
days) during the third to fifth year after men- diagnosis is delayed puberty (some consider
arche. Abnormal uterine bleeding occurs when boys delayed if no signs appear by 13.5 years).
Abnormalities of Puberty 203

Table 9.3 Differential diagnosis of delayed puberty


Serum Serum gonadal
Condition gonadotropins steroids Miscellaneous
Constitutional delay Prepubertal Low Patient usually has short stature for chronologic age but
in growth and (low) appropriate height and growth rate for bone age. Both
adolescence adrenarche and gonadarche are delayed.
Hypogonadotropic Prepubertal Low Patient may have anosmia (Kallmann syndrome) or other
hypogonadism (low) associated pituitary hormone deficiencies. If gonadotropin
deficiency is isolated, the patient usually has normal height
and growth rate but lacks a pubertal growth spurt.
Adrenarche may occur at a normal stage in spite of absent
gonadarche (serum DHEAS may be pubertal).
Hypergonadotropic Elevated Low Patient may have abnormal karyotype and stigmas of
hypogonadism Turner syndrome or Klinefelter syndrome.

This can be an example of normal variation, such delay, although the rule is not constant for all.
as constitutional delay in puberty, or a serious Unlike patients with Kallmann syndrome
condition, such as gonadotropin deficiency due to (below), the sense of smell is normal and there is
a central nervous system tumor. Thus the etiol- no increased incidence of cryptorchidism nor
ogy of delayed puberty is always worth serious microphallus. The condition has a strong family
consideration. The clinician must determine pattern as 50–75 % of patients have a family
which patient and parents need reassurance and history of the same.
which need testing to determine if a serious con- There is no definitive diagnostic test to estab-
dition is present (Table 9.3). lish constitutional delay or to separate it from
hypogonadotropic hypogonadism although the
Constitutional Delay in Puberty finding of a value of inhibin B >35 pg/mL in one
(See Chap. 5) study supported the diagnosis of CDP while in
CDP is usually accompanied by constitutional another study inhibin B <111 pg/mL with basal
delay in growth in childhood. These are children, LH <0.3 IU/L was highly diagnostic for
more frequently boys, who tend to be thin and IHH. Standards for the inhibin assay vary with
who have a physiologic delay in all measurable the method and laboratory, so a distinct cutoff is
aspects of growth, but not in mental develop- not yet established in this diagnostic situation.
ment. Thus, CBC, alkaline phosphatase, and Clearly the history and physical findings must
other biological measures that change with be considered as a whole and elimination of
pubertal development are characteristic of a other pathologic conditions must occur before
young child. The bone age is delayed as well, and accepting the diagnosis; this is a diagnosis of
the height age (the height age is the chronologic exclusion.
age at which the child’s height is equal to the 50th
percentile on the growth chart) is delayed for age Hypogonadotropic Hypogonadism
but generally appropriate for the bone age (the (Fig. 9.2b) (#146110)
bone age has significant limitations in accuracy, Hypogonadotropic hypogonadism (HH) may be
however). These children have plasma gonado- due to a genetic defect in gonadotropin secretion, a
tropin and sex steroid values characteristic of developmental defect, a tumor, or other CNS abnor-
younger children and a phenotype more appro- malities. Individuals will characteristically grow
priate for a younger child. Sometimes watchful normally at first in isolated hypogonadotropic
waiting is necessary for years after the first evalu- hypogonadism (IHH), but will have no pubertal
ation until pubertal development proceeds in the growth spurt due to the lack of the rising sex ste-
natural history of the condition, but almost all roids in puberty. If growth hormone is affected as
subjects begin pubertal development by 18 well as gonadotropin secretion with multiple pitu-
years of age. Delayed adrenarche and gonad- itary deficiencies, the child will have a decreased
arche are often found together in constitutional growth rate during childhood and puberty.
204 9 Disorders of Puberty

Isolated Hypogonadotropic the KAL1 gene leads to abnormalities in the pro-


Hypogonadism duction of anosmin, which is a neural cell adhe-
Patients with IHH characteristically grow normally sion molecule that is responsible for this normal
during childhood but, with the absent pubertal migration of the involved cells. Autosomal domi-
growth spurt, fall behind the height of typical nant Kallmann syndrome 2 (*147950, KAL2) is
subjects. Since they have a delay in epiphyseal associated with defects of the midline such as
fusion, they may continue to grow to a taller adult cleft palate, neurosensory hearing loss, and
height. The individual may develop eunuchoid extremity movements known as synkinesia; it is
proportions consisting of long legs and arms and due to mutation in the fibroblast growth factor
an upper-to-lower segment ratio well below 0.9, receptor 1 gene (FGR1 gene). Kallmann syn-
the value ordinarily normal for adults. IHH may drome 3 (*244200; KAL3) manifests with mid-
be a congenital defect, sometimes related to a line defects such as cleft palate or lip as well as
defined mutation. However, acquired gonadotro- unilateral renal agenesis; it is due to a mutation in
pin deficiency is an ominous sign and could be an the G protein-coupled prokineticin receptor-2
indication of a central nervous system tumor. gene or its receptor and is thought to be an autoso-
There is a continuum in the manifestation of mal dominant condition. The CHARGE syn-
gonadotropin deficiency ranging from complete drome (#214800 ICD+CHARGE SYNDROME
absence of puberty to delay in puberty extending consisting of colobomata, heart anomalies, cho-
to reproductive defects alone. Most recently, it anal atresia, retardation, genital and ear anoma-
was determined that 10–20 % of patients with lies) includes IHH and hyposmia and is caused by
proven genetic defects who would be expected to mutations in the CHD7 gene and is inherited in an
have permanent hypogonadotropic hypogonad- autosomal dominant pattern; patients with
ism can recover function and demonstrate normal CHARGE syndrome should be evaluated for
reproductive hormone secretion; thus continued hypogonadotropic hypogonadism, especially if
evaluation is required after the initial diagnosis is there is deafness and hypoplasia of the semicircu-
made. Within any given family, there may be indi- lar canals. An ever-increasing number of genes are
viduals with constitutional delay and other rela- associated with KS and IHH in an oligogenetic
tions with Kallmann syndrome demonstrating the manner indicating that inheritance occurs in a
overlapping nature of conditions causing hypogo- more complicated manner than in simple
nadotropic hypogonadism. Mendelian patterns; these oligogenic patterns may
explain why several affected members of the same
Kallmann Syndrome (KS) family may have different clinical characteristics.
KS is the most common genetic form of IHH. KS Loss-of-function mutations of the KISS1R
in the classic form manifests with an impaired previously known as GRP54 (#614837
sense of smell (hyposmia or anosmia) and gonad- HYPOGONADOTROPIC HYPOGONADISM 8
otropin deficiency. However, there is great genetic WITH OR WITHOUT ANOSMIA; HH8), the G
heterogeneity, and within a given family, some protein-coupled receptor 54 for kisspeptin, lead to
patients may have a normal sense of smell with no hypothalamic hypogonadism. Although rare, these
reproductive hormone function, while others may patients have been instrumental in understanding
have a normal sense of smell with reproductive the kisspeptin–GnRH axis. Loss-of-function
ability. The classic X-linked Kallmann syndrome mutations in the GNRH-receptor gene have been
(308700 Kallmann syndrome 1; Kal1 at Xp22.3) noted for several years (#228300 FERTILE
is due to impaired migration of GnRH neurons EUNUCH SYNDROME), but more recently
from the anterior region of the embryonic nose mutations in the GNRH gene (#614841
(the olfactory or cribriform plate) to the normal HYPOGONADOTROPIC HYPOGONADISM
location of GnRH neurons in the medial basal 12 WITH OR WITHOUT ANOSMIA; HH12)
hypothalamus, above the pituitary portal system, have been discovered and linked to hypothalamic
by about 20 weeks of gestation. The mutation of hypogonadism. Mutations of the DAX gene
Abnormalities of Puberty 205

(#300200 ICD+ADRENAL HYPOPLASIA, indicate a tumor. If both anterior and posterior


CONGENITAL; AHC) are associated with hypo- pituitary deficiencies are noted, a central nervous
gonadotropic hypogonadism, adrenal hypoplasia, system tumor should be even more highly
glycerol kinase deficiency, and muscular dystro- suspected.
phy. Normally, there are beta and alpha subunits To reemphasize, late onset of growth failure
to the FSH and LH molecules, the alpha subunit or a cessation of pubertal development that has
being common to all pituitary-glycoprotein already started is of great concern.
hormones while the beta subunit confirms speci-
ficity of action. Rarely mutations of FSH beta Autoimmune or Lymphocytic
gene (#229070 FOLLICLE-STIMULATING Hypophysitis
HORMONE DEFICIENCY, ISOLATED) or Autoimmune hypophysitis is rarely reported in
mutations of the LH beta gene (*152780 lutein- childhood. It may involve lymphocytic infiltra-
izing hormone, beta-polypeptide) are reported in tion of the pituitary gland. It presents as a pitu-
which there is no production of intact FSH or LH itary mass associated with hypopituitarism.
molecules. Without the FSH beta gene, sper- Hypogonadotropic hypogonadism is one possi-
matogenesis cannot progress while without the ble outcome.
LH beta gene, Leydig cell stimulation will not Craniopharyngiomas are tumors of Rathke’s
occur and the patient may have the phenotype of pouch originating in the pituitary stalk. Spread to
the “fertile eunuch syndrome” in which there is the suprasellar region or even the sella turcica is
lack of physical evidence of testosterone action possible. Peak incidence is between 6 and 14
while the seminiferous tubules enlarge and sper- years of age. Patients may be symptomatic with
matozoa can develop. complaints of headache, visual loss, polyuria,
Other mutations more rarely causing hypotha- and polydipsia. Signs on physical exam may be
lamic hypogonadism without anosmia include the either short stature or cessation of previously nor-
steroidogenic factor 1 (SF1) as well as TAC3, mal growth, findings of hypothyroidism, and
TACR3, CHD7, and NELF. Commercial labora- sexual infantilism. Papilledema or optic atrophy
tories are now able to probe for mutations in many (not congenital optic dysplasia) might be found
of these genes. as well. On occasion, slow-developing cranio-
Mutations in the LH receptor manifest in a pharyngiomas are diagnosed during evaluation of
phenotypic male with micropenis, or ambiguous other conditions. For instance, pituitary calcifica-
genitalia, or even an XY phenotypic female tions may be found on a radiograph taken during
(#238320 ICD+LEYDIG CELL HYPOPLASIA, orthodontic evaluation. CT scans will show flecks
TYPE I); this is discussed in Chap. 8. of calcium within the tuber cinereum in more
than 80 % of cases. MRI scans before and after
Abnormalities of the Central Nervous gadolinium will define the shape and size of the
System tumor better although it will not show calcifica-
These conditions are prime examples of why the tion without modification. This tumor might be
diagnosis of the etiology of delayed puberty must cystic, and the sella turcica may be eroded by
be carefully established; if untreated, many of expansion of the tumor. Transsphenoidal micro-
these conditions can be fatal. surgery can be used for extirpation if the tumor
is intracellular. Larger tumors that cannot be
Central Nervous System Tumors completely removed by this method may be
(Many of these are discussed in Chap. 3) treated with craniotomy and radiation as the
The secretion of any hypothalamic pituitary tumors are radiosensitive. However, late effects of
hormone can be affected by tumors of the CNS. radiation are possible with CNS irradiation, such
Thus late onset of gonadotropin deficiency, as as secondary tumors of local tissue.
opposed to congenital gonadotropin deficiency, Other tumors of the central nervous system
is an ominous sign since the clinical findings may reside outside of the sella turcica such as
206 9 Disorders of Puberty

germinomas of the pineal, which by their mass (#1182238 septo-optic dysplasia). SOD is com-
may lead to HH; since they secrete hCG which patible with normal pituitary function in some
acts as LH, they may cause peripheral precocious patients, but deficiencies should be considered in
puberty in males but not females. Astrocytomas all. SOD can also cause CPP.
and gliomas (which may or may not be associ- Other midline defects of the central nervous
ated with neurofibromatosis) are other CNS system associated with pituitary deficiencies
tumors that may cause HH. range from a simple cleft palate (6 % have growth
Intracellular adenomas are rare, but they may hormone deficiency) up to holoprosencephaly.
impair pituitary function. Most do not secrete
hormones in childhood, but prolactinomas, which Hypogonadotropic Hypogonadism
are rare in childhood, will manifest galactorrhea Following Irradiation
as a diagnostic characteristic of this prolactin– Oncofertility is a new field growing out of the
secreting tumor. Hyperprolactinemia may delay increasing number of cancer survivors with
puberty or cause amenorrhea. infertility following successful treatment of their
Histiocytosis X may be associated with diabe- cancer. Radiation treatment may cause growth
tes insipidus, but other hypothalamic hormones hormone deficiency or gonadotropin deficiency
may also be deficient and HH can occur. but also, when used to treat a central nervous sys-
Granulomas of tuberculosis sarcoidosis, postin- tem tumor, may lead to a combination of preco-
fectious inflammation, or vascular lesions of the cious puberty and growth hormone deficiency.
central nervous system can impair hypothalamic The growth hormone deficiency will not be dem-
pituitary function. CNS trauma due to accidents, onstrated by extremely slow growth if the puberty
child abuse, or surgery for tumors near the area is progressing rapidly as the sex steroid secretion
can also affect hypothalamic pituitary gonadal stimulates growth masking the GH deficiency
function. Hydrocephalus can cause GnRH defi- (read below).
ciency as may any other etiology of increased
intracranial pressure, such as a subarachnoid cyst. Idiopathic Hypopituitarism
The most frequent pituitary hormone defect with
Congenital Defects of the CNS hypothalamic deficiency is growth hormone defi-
(See Chap. 3) ciency, but gonadotropin deficiency follows in
Congenital defects of the CNS are characteristi- prevalence. If both growth hormone and gonado-
cally found in the neonatal period or soon there- tropin function are deficient, accurate diagnosis
after. They may be associated with single may be difficult. Growth hormone deficiency
hypothalamic–pituitary defects or more com- itself can delay puberty which, of course, would
monly multiple deficiencies. also be found in gonadotropin deficiency. While
hypopituitarism is usually sporadic, familial
Septo-optic Dysplasia hypopituitarism may be found in X-linked
Septo-optic dysplasia (SOD), or the appearance (*3120000 panhypopituitarism) and autosomal
of small, pale optic discs, is associated with recessive pattern (#262600 pituitary dwarfism
impaired vision ranging from mild defects to III) and may be found with diabetes insipidus
blindness and is often found with horizontal or (241540 hypopituitarism, congenital, with cen-
pendular (to and fro) nystagmus. Optic hypopla- tral diabetes insipidus). Mutation in the pituitary
sia is to be differentiated from optic atrophy paired-like homeodomain transcription factor
which may be an ominous sign of a tumor causing (PROP1 #262600 PITUITARY HORMONE
the degeneration of a previously normally formed DEFICIENCY, COMBINED, 2; CPHD2) causes
optic disc. About 50 % of patients with optic dys- hypopituitarism with absence of PRL, LH, FSH,
plasia have absence of the septum pellucidum GH, or TSH. However, patients with mutations of
and a diagnosis of SOD. SOD may be the result another related transcription factor
of a mutation in the homeobox gene HESX POU1 (#613038 PITUITARY HORMONE
Abnormalities of Puberty 207

DEFICIENCY, COMBINED, 1; CPHD1) lack SNRPN gene may be found in other affected
GH, prolactin, and TSH but retain ACTH and patients (see growth Chap. 5 for more).
gonadotropin function (ACTH deficiency may Laurence–Moon syndrome (*245800
develop later however). Laurence–Moon) and Bardet–Biedl (#209900
Boys with congenital hypopituitarism might Bardet–Biedl syndrome) syndrome share some
present at first with microphallus, a normal- features and were considered separate entities
appearing penis which, however, is less than but are now combined into the Bardet–Biedl
2 cm (2.5 cm in some sources) in stretched syndrome with an autosomal recessive pattern
length at birth. Associated growth hormone or of inheritance. Obesity, short stature, develop-
ACTH deficiencies might occur and lead to mental delay, and retinitis pigmentosum can be
hypoglycemic seizures. Thus, hypoglycemia and associated with hypogonadotropic hypogonad-
microphallus in a newborn boy should cause ism or hypergonadotropic hypogonadism as
consideration of congenital hypopituitarism, well. Spastic paraplegia, polydactyly, and other
especially if pendular nystagmus occurs in addi- features noted in Chap. 5 are found.
tion as an indication of SOD. Microphallus can
be treated with low-dose testosterone therapy Weight Loss and Chronic Disease
(25 mg intramuscularly of testosterone enan- Weight loss due to chronic disease, malnutrition,
thate every month for three doses) to enlarge the and even dieting to less than 80 % of ideal weight
penis without causing undue advancement of the can cause hypogonadotropic hypogonadism. A host
bone age. Sex reversal should not be considered of diseases affecting nutrition including poorly
in boys with congenital hypopituitarism and treated diabetes mellitus, gastrointestinal disorders
microphallus since the penis will grow with tes- causing malabsorption or celiac disease, connec-
tosterone therapy and the outcome is good. The tive tissue disorders, among others can cause
clinician must be aware that, later in life, obese delayed puberty. Anorexia nervosa is a psychiatric
boys are often referred for microphallus when, in disorder characterized by a distorted body image,
fact, their penis is normal in width as well as obsessive fear of obesity, and food avoidance that
length but is buried in fat; in these cases, the width can cause severe self-induced weight loss, primary
of the penis should be a good indication of whether or secondary amenorrhea in affected females,
the penis is normal even if it is mostly hidden. widespread endocrine disorders, and even death as
Females with congenital hypopituitarism have this condition has a higher mortality rate than any
normal genitalia at birth but like boys may have other psychiatric diagnosis (mortality rate is approx-
pendular nystagmus and hypoglycemic seizures. imately 15 %). Specific diagnostic details are pro-
vided in the Diagnostic and Statistical Manual of
Syndromes Associated Mental Disorders, fifth edition [DSM-V], criteria
with Hypogonadotropic Hypogonadism of the American Psychiatric Association. The onset
Prader–Willi syndrome (#176270) includes fetal of puberty may be delayed in the most seriously
and infantile hypotonia, short stature, obesity, affected younger subjects. Osteopenia or osteoporo-
lack of satiety, as well as almond-shaped eyes sis is a consequence of estrogen deficiency. Weight
and characteristic facies. Hands and feet are loss causes reversion of gonadotropin secretion pat-
small (acromicia). The children are developmen- terns to prepubertal low-amplitude pulsatile secre-
tally delayed, and microphallus and undescended tion and many months may pass before hormonal
testes in boys and delayed menarche in girls are function is regained with weight gain. Even after
often found. While this is usually associated with weight gain, however, amenorrhea may persist, pos-
short stature, some children are normal or tall in sibly related to the psychiatric disease itself.
childhood. The etiology of this condition is a Bulimia nervosa is a syndrome of binge eating
microdeletion of 15q11 in about 70 % of patients and purging. Evidence of this condition, besides
which can be confirmed with fluorescent in situ purging behaviors, includes frequent trips to the
hybridization (FISH); maternal disomy of the bathroom after meals, evidence of vomiting in
208 9 Disorders of Puberty

the presence of laxative, or diuretic packaging in causing disorders of sexual differentiation are
personal spaces like under beds. Affected patients also discussed in Chap. 6.
often have severe, rigid exercise episodes.
Vomiting may lead to unusual swelling of the Disorders of the Ovaries
cheeks and jaw area due to parotid irritation and
there may be calluses on the palms (Russell sign) Turner Syndrome
or knuckles as evidence of self-induced vomit- The Turner syndrome of gonadal dysgenesis is
ing. Teeth may be stained from the effects of the the most common form of primary gonadal fail-
acid in the vomitus. Growth may be impaired and ure in a female phenotype; the prevalence is
bone density may be decreased. Like anorexia between 1:2,000 and 1:5,000 live phenotypic
nervosa this syndrome can be fatal. female births. Turner syndrome is caused by a
The female athlete triad incorporates the fea- loss or abnormality of one X chromosome or a
tures of decreased bone density and menstrual portion of it. It is estimated that the 45X karyo-
abnormalities with energy intake out of balance type occurs in 1 of 15 spontaneous abortions and
with energy expenditure; recent recommenda- that 99.9 % of 45X fetuses do not survive longer
tions suggest referring instead to “Relative than 28 weeks of gestation. Classic cases of
Energy Deficiency in Sport” (RED-S) which Turner syndrome have a 45X karyotype. There is
encompasses the condition in boys as well and no Barr body found on buccal smear after Giemsa
directs attention to the basic etiology. These girls staining (this test has been replaced by karyotype
are characteristically thin; however, athletic determination; Barr body determination should
amenorrhea may also occur in heavier girls who no longer be used for diagnosis because of lack
exercise excessively when athletic amenorrhea is of reliability). Physical features of Turner syn-
found in swimmers or ice skaters. Decreased drome include short stature (average adult height
activity in these heavier girls allows the resump- is 143 cm), streak gonads (a normal complement
tion of normal pubertal development and menses of ova is present at birth, but accelerated oocyte
without necessarily an increase in weight. apoptosis occurs in the postnatal period), sexual
Hypothyroidism will inhibit the onset of infantilism, and a female phenotype. Other find-
puberty and menses or, if it occurs after the onset ings include down-turned edges of the mouth
of puberty, will stop the progression of puberty. (leading to a “fish-mouth appearance”) with ret-
Primary hypothyroidism may be associated with rognathia and high-arched palate, ptosis and epi-
galactorrhea due to elevated prolactin as well as canthal folds, broad shield-like chest, the
elevated TSH, both due to increased TRF secre- appearance of wide-spaced and hypoplastic
tion. Sometimes, severe primary hypothyroidism poorly pigmented nipples, short webbed neck
is associated with some features of precocious with low hairline, short fourth metacarpals,
puberty in the Van Wyk–Grumbach syndrome Madelung abnormality of the wrist (radial short-
due to stimulation of the gonads by extremely ening and bowing with dorsal subluxation of the
high TSH values which allows cross-reaction distal ulna and limited rotation), wide-carrying
with gonadotropin receptors. angle of the arms (cubitus valgus), knock-knee
appearance (genu valgum), multiple nevi, spoon-
Hypergonadotropic Hypogonadism shaped (hyperconvex) hypoplastic nails, and
(Fig. 9.2c) lymphedema of the extremities, particularly in
Hypergonadotropic hypogonadism is due to infancy. At birth, many patients have lymph-
primary gonadal failure. Serum gonadotropins edema of the extremities and loose skin folds
are elevated because of lack of feedback inhibi- around the neck, which later transform into the
tion from steroid or inhibin secretion from the classic webbed neck and also account for the
abnormal gonads. Most patients have a karyo- low-set ears; this newborn appearance of new-
type abnormality or a history of prior chemo- born Turner syndrome is the Bonnevie–Ullrich
therapy or radiotherapy. The congenital defects syndrome. A tendency exists toward keloid
Abnormalities of Puberty 209

formation, which can complicate attempts to XO/XX). Such patients may have some gonadal
correct the webbed neck surgically. A girl with function and a more typical female phenotype.
Turner syndrome may not manifest all the physi- Other variants of Turner syndrome include
cal findings and may appear as an average short patients with mosaicism involving the Y chromo-
girl, so a high index of suspicion in any short girl some, such as XO/XY. A FISH probe for Y chro-
without a diagnosis is indicated; rarely girls with mosome material should be performed if this
taller familial heights will not even be short and situation is suspected. These patients may have
hypogonadism will be the indicator. Turner syn- dysgenetic testes rather than streak gonads and
drome is considered a gonadal disorder of sexual are at risk for the development of gonadoblastoma
differentiation (DSD). or malignant degeneration of the gonad, so gonad-
The SHOX gene (*312865 SHORT STATURE ectomy is indicated. Gonadoblastomas may
HOMEOBOX; SHOX at Xpter-p22.32) on the secrete estrogen falsely suggesting residual ovar-
short arm of the X chromosome near the pseudo- ian function. Phenotype may be infantile female
autosomal region is absent on the aberrant or to ambiguous genitalia or phenotypic male.
absent X chromosome; this gene is related to the Growth hormone (GH) treatment is approved
short stature of Turner syndrome and gives rise to for Turner syndrome; treatment increases stature in
the Madelung deformity of the wrist; absence of Turner syndrome above the average adult height of
the SHOX gene is a cause of short stature in other 143 cm often to 152 cm or more. The age to initiate
children without Turner syndrome (see Chap. 5). estrogen replacement is controversial; if it is
The genes determining ovarian development are offered too early and in high dosage, it will
also located on the X chromosome. In addition to decrease the adult height, while late start may
the abnormalities of the wrist, girls with Turner intensify psychological problems. Recent evidence
syndrome are predisposed toward kyphoscoliosis shows that low doses of estrogen offered at the
and osteopenia. average age of puberty will not affect height when
Patients with the 45X karyotype usually have used in conjunction with GH (see growth Chap. 5).
normal intelligence but may perform poorly on
tests of visual–spatial perception and demonstrate Other Forms of Primary Ovarian Failure
other defects in memory and executive function; Ovarian failure can occur with chemotherapy for
other karyotypes may be associated with defects in malignant disease. If the ovaries are not shielded
intelligence. Frequent episodes of otitis media in or surgically moved out of the path of a beam of
childhood may lead to conductive hearing loss, abdominal/pelvic radiation therapy, failure may
although sensorineural hearing loss is possible, as occur.
well. Abnormalities of the shape of the kidneys FSH receptor mutations (#233300 OVARIAN
(e.g., horseshoe kidneys or duplications) may lead DYSGENESIS 1; ODG1) can occur in a pheno-
to frequent urinary tract infections in those with typically female individual who does not develop
abnormal kidney function, so ultrasound evalua- secondary sexual characteristics at puberty and
tion of kidneys is indicated. Left-sided heart anom- does not menstruate. There are bilateral “streak
alies (such as coarctation of the aorta often first gonads,” consisting of fibrous tissue and variable
noted due to hypertension soon after birth or bicus- amounts of wavy ovarian stroma. Affected
pid aortic valve) are common. Affected girls females have a uterus and fallopian tubes and
require cardiac evaluation for heart defects and external genitalia are infantile female.
appropriate follow-up care. Even normotensive Galactosemia (*606999 GALACTOSE-1-
patients require regular screening for aortic dilation PHOSPHATE URIDYLYLTRANSFERASE;
every 5–10 years as adults. Increased incidence of GALT) presents after neonatal galactose inges-
Hashimoto thyroiditis, celiac disease, type 1 diabe- tion with jaundice, hepatosplenomegaly, hepato-
tes, and insulin insensitivity are other features. cellular insufficiency, hypoglycemia, renal
Variants of Turner syndrome occur with tubular dysfunction, muscle hypotonia, sepsis,
abnormal X chromosome or mosaicism (e.g., tendency toward E. coli sepsis, growth failure,
210 9 Disorders of Puberty

feeding dysfunction, and cataract. Long-term or absence of the uterine cervix in the presence of a
complications include mental retardation, verbal functional uterus may present in this way.
dyspraxia, motor abnormalities, and hypergonad-
otropic hypogonadism in females only. 46,XY and XX DSD
Galactosemia screening is performed at birth in 46,XY disorder of sexual development (DSD),
most countries. previously called male pseudohermaphroditism,
Premature menopause has also been described in is an alternative cause of primary amenorrhea if a
otherwise healthy girls owing to the presence of anti- patient has achieved thelarche. The syndrome of
ovarian antibodies. Patients with Addison disease complete androgen resistance leads to female
may have autoimmune oophoritis as well as adrenal external genitalia and phenotype without axillary
failure. Polyglandular autoimmune syndromes 1 or pubic hair development in the presence of
(#240300 AUTOIMMUNE POLYENDOCRINE pubertal breast development (androgen resis-
SYNDROME, TYPE I; APS1) and 2 (%269200 tance or the syndrome of testicular feminization;
AUTOIMMUNE POLYENDOCRINE see Chap. 8 and below).
SYNDROME, TYPE II; APS2) combine various XX and XY gonadal dysgenesis may be spo-
auto endocrine organ failure and premature ovarian radic or familial. Stature is normal, and pheno-
failure can occur as well. type is sexually infantile female in the XX form
A sex steroid biosynthetic defect due to 17-alpha- (*233400 GONADAL DYSGENESIS, XX
hydroxylase deficiency (P450c17) (#202110 TYPE, WITH DEAFNESS) or may be ambigu-
ADRENAL HYPERPLASIA, CONGENITAL, ous in the XY form (*306100 GONADAL
DUE TO 17-ALPHA-HYDROXYLASE DYSGENESIS, XY FEMALE TYPE; GDXY).
DEFICIENCY) is manifested as sexual infantilism A FISH probe for Y chromosome material
and primary amenorrhea in a phenotypic female should be performed if this situation is sus-
(regardless of genotype since testes and ovaries can- pected. Patients with an XY karyotype should
not produce sex steroids) with hypertension and undergo gonadectomy because of potential neo-
hypokalemic alkalosis (see Chap. 8 on sexual dif- plastic degeneration of the dysgenetic testes
ferentiation). Carriers of the FMR1 gene responsi- (see Chap. 8).
ble for fragile X syndrome are susceptible to
premature ovarian failure. Disorders of the Testes

Infectious Oophoritis Klinefelter Syndrome


Infectious oophoritis may be caused by gonor- Klinefelter syndrome, or seminiferous tubular
rhea or chlamydia. This is an ascending infection dysgenesis, is the most common cause of testicular
from the lower reproductive tract. failure, with a prevalence of 1 in 500–1,000 males.
Because of variable Leydig cell function, serum
Primary Amenorrhea Associated testosterone levels in the adult patients vary from
with Normal Secondary Sexual low to close to normal. Therefore, the age of onset
Development of puberty may be normal, but secondary sexual
A structural anomaly of the uterus or vagina changes do not progress to the adult stage. In
interfering with the onset of menses in spite of puberty or in the adult, testes are firm and grow no
normal endocrine milieu will present with pri- longer than 3.5 cm, with histologic changes of
mary amenorrhea in the presence of normal hyalinization and fibrosis of the seminiferous
breast and pubic hair development. A transverse tubules progressing with pubertal development.
vaginal septum, an imperforate hymen, or the Seminiferous tubular function is invariably
Rokitansky–Kuster–Hauser syndrome (%277000 affected, and impaired spermatogenesis is the rule;
MAYER–ROKITANSKY–KUSTER–HAUSER microsurgery may allow the removal of a few
SYNDROME) of congenital absence of the vagina remaining normal spermatozoa to allow assisted
combined with abnormal development of the uterus fertility in adults.
Abnormalities of Puberty 211

Even before the onset of puberty, testes are 46,XY DSD


small and the upper-to-lower segment ratio is LH receptor mutation leads to lack of testosterone
decreased for age, although arm span is not production. Two types are noted. Type I (#238320
greater than height as is found in patients with LEYDIG CELL HYPOPLASIA, TYPE I and II),
eunuchoid proportions, and these body propor- complete inactivation of LHCGR, demonstrates
tions persist into adulthood. Stature is often taller complete 46,XY DSD (formerly male pseudoher-
than average in Klinefelter syndrome. maphroditism), low testosterone and high LH lev-
Serum gonadotropin concentrations may be els, total lack of responsiveness to LH/CG challenge,
measured at the upper limit of the normal range phenotype of an immature female with lack of
or slightly above this limit during childhood. breast development, and absent development of
Serum LH and especially FSH are elevated to secondary male sex characteristics. Type II, due to
castrate levels after pubertal age due to decreased partial inactivation of the gene, leads to patients in a
testosterone and/or inhibin production from the spectrum from micropenis to severe hypospadias.
affected testes. Gynecomastia is common, and Females with inactivating mutations in the LHCGR
breast cancer may develop (a 20-fold increased gene display defective follicular development and
risk over a nonaffected male). Germ cell tumors ovulation, amenorrhea, and infertility.
of the mediastinum also are more likely in Complete androgen insensitivity syndrome
affected males. (#300068 ANDROGEN INSENSITIVITY
Some patients come to evaluation because of SYNDROME; AIS) presents as a 46,XY pheno-
personality disorders and learning disability in typic female with amenorrhea and absence of
the prepubertal age range. A verbal IQ dispropor- pubic and axillary hair. This condition follows an
tionately lower than performance IQ and expres- X-linked inheritance pattern. Partial androgen
sive developmental defects in speech and insensitivity syndrome (#312300 ANDROGEN
language are found. A tendency toward explosive INSENSITIVITY, PARTIAL; PAIS) manifests as
outbursts and difficulty in controlling anger are 46,XY DSD with 46,XY karyotype, ambiguous
seen, even though the individuals otherwise tend genitalia with hypospadias, hypogonadism,
to be passive. Thus the diagnosis should always gynecomastia, and infertility in an X-linked
be entertained in a child with any of these person- recessive pattern.
ality or learning problems; since karyotype deter- Noonan syndrome (#163950 NOONAN
mination is often invoked in the diagnosis of SYNDROME 1; NS1 12q24.1) (previously termed
learning difficulties, the diagnosis of Klinefelter pseudo-Turner syndrome or male Turner syn-
syndrome is occurring at younger and younger drome) is a dominantly inherited condition which
ages. Further, the use of amniocentesis for other has no biological relationship to Turner syndrome
indications allows prenatal diagnosis of in spite of sharing some physical features with
Klinefelter syndrome as well. Turner syndrome (such as webbed neck, ptosis,
Klinefelter syndrome results from nondisjunc- short stature, wide-carrying angle, and lymph-
tion of the sex chromosomes. It is more common edema), as well as features different from those of
with advanced maternal age. The karyotype is clas- Turner syndrome (such as normal karyotype, trian-
sically 47,XXY; we do not recommend obtaining a gle-shaped face, pectus excavatum, right-sided
buccal smear, although it would be positive in con- heart disease in contrast to left-sided heart disease
trast to a 46,XY male in which a buccal smear is in Turner syndrome, and, more commonly, devel-
negative. Variants of Klinefelter syndrome are opmental delay). Affected boys may have unde-
reported, with mosaicism such as in XX/XXY, scended, often impaired, testes. Growth hormone is
XXYY, XXXY, and XXXXY karyotypes. Other approved for treatment of short stature in this con-
patients are reported with features similar to those dition (see growth Chap. 5). Genetic diagnosis is
of Klinefelter syndrome but with an XX karyotype available with mutations at PTN11 mutations on
(XX males); in these cases, transfer of the SRY 12q24.1 found in some patients. Some reports link
from the Y to the X chromosome is the cause this mutation with a decreased response to GH but
(see Chap. 8 on sexual differentiation). recent reports question this finding.
212 9 Disorders of Puberty

Radiation and Chemotherapy Damage It is important to differentiate bilateral crypt-


Survival is now possible in a wide range of child- orchidism from anorchia. In anorchia, the testes
hood cancers. However, radiation therapy to the were present during fetal life to allow the devel-
area of the gonads or chemotherapy may dimin- opment of a normal male phenotype including a
ish or limit gonadal function. Elevated gonado- normal penis and normal Wolffian duct struc-
tropins suggest the diagnosis of gonadal failure, tures, but the testes atrophied, possibly because
and this may be noted only after a period of fol- of vascular insufficiency subsequent to genital
low-up. Methods to preserve fertility in children development leaving the scrotum small and
treated for cancer are under investigation in the “uninhibited.” A sequence of evaluations may
field of oncofertility and some show promise for determine whether bilateral cryptorchidism or
future fertility. Recent concepts consider quality anorchia is present. Ultrasound examination in
of remaining germ cells rather than quantity. experienced hands can reveal the presence of the
Spermatogonia have been recovered from radia- testes, but lack of visualization may not be defini-
tion- or chemotherapy-damaged testes or ovaries. tive proof of absence of the testes. Elevation of
gonadotropins above the normal for age indicates
Anorchia and Cryptorchidism lack of functioning of testicular tissue. Detection
Cryptorchidism refers to unilateral or bilateral of inhibin or AMF indicates the presence of tes-
undescended testes and must be differentiated ticular tissue and can help to eliminate the diag-
from anorchia, which is the complete absence of nosis of anorchia; inhibin B values more than 3
both testes in a phenontypic male. Anorchia is standard deviations below the mean are compati-
also known as the testicular regression syndrome. ble with anorchia.
While the testes develop in the abdominal cavity, Stimulation tests have been evaluated for use
by month 7 of gestation they usually descend into in cryptorchidism in Prader–Willi syndrome; the
the scrotum. The peptide hormone insulin-like 3 protocol may be repeated in a patient with bilat-
(INSL3) is expressed at high levels in fetal Leydig eral undescended testes without Prader–Willi
cells and interacts with the relaxin family peptide syndrome. The administration of a single dose of
receptor 2 (RXFP2) to promote the testicular 1,500 IU i.m. hCG is administered in one diag-
descent. INSL3 also plays a role in adult fertility. nostic schema for the Prader–Willi syndrome,
More than 97 % of term and 79 % of premature and baseline and 72 h afterwards serum levels of
infant boys have descent of the testes. By age 9 LH, FSH, testosterone, inhibin B, and AMH are
months, 99.2 % of testes are descended, with little measured. The result is normal when the maxi-
progress thereafter. Thus by age 9–12 months, mum testosterone level after 72 h is 2–20 times
most testes that will descend spontaneously will higher than baseline in infants aged 3–12 months
have done so. Lack of descent is associated with a and 5–10 times higher, between 2.5 and
tenfold increase in neoplasia, and the inability to 9.0 nmol/L, in infants aged 1–4 years. If no
examine undescended testes for neoplastic increase in testosterone can be induced with one
changes makes the situation worrisome. Thus, by to six injections of hCG, the patient may have no
age 1 year, all undescended testes should undergo Leydig cells and therefore has anorchia.
orchiopexy or, if abnormal, orchiectomy. Although There are studies of longer-term administra-
an increase in the incidence of carcinoma is noted tion of hCG or GnRH to foster descent of testes
if the testes are left in their undescended position, in boys with variable results. Administration of
there is evidence that undescended testes have an an hCG dose, 250 IU for infants aged 3–12
intrinsic problem, as one-third of carcinomas months of age and 500 IU IM for infants aged
develop in cryptorchid testes after orchiopexy, and 1–4 years, twice a week for 6 weeks led to a
20 % of cryptorchid patients have carcinoma in descent of testes in 62 % who had a positive stim-
the contralateral descended testes. Unilateral ulation test and 23 % of the testes reached a sta-
anorchia may lead to compensatory hypertrophy ble scrotal position. GnRH or GnRH agonist has
of the contralateral testes. been used to promote testicular descent in some
Abnormalities of Puberty 213

studies. It has been suggested that hCG and of bacteria and viruses. The most common cause
GnRH caused descent of undescended testes only of orchitis has been a viral infection with the
in cases of retractable or “yo-yo” testes, which mumps virus. It most often occurs in boys after
travel up and down in the canal at various times puberty. Orchitis usually develops 4–6 days after
and there is no situation in which surgery might the mumps begins. Because of childhood vacci-
not be indicated in the first place. nations, mumps is now rare in the USA. Orchitis
If no indication of testicular function is found may also occur because of infections with sexu-
and then if no descent of the testes can be brought ally transmitted diseases including gonorrhea and
about, the question of exploratory surgery arises. Chlamydia.
Exploratory surgery, which may lead either to
orchiopexy if normal testicular tissue is found or Differential Diagnosis of Delayed
to a diagnosis of anorchia if no testes are found, Puberty (Fig. 9.3)
may be indicated. However, a recent study stated If the onset of puberty is delayed according to
that there is a high degree of concordance of the guidelines above, determination as to whether
endocrine testing indicating anorchia with surgi- the patient has primary gonadal disease and
cal findings and suggested that exploratory sur- hypergonadotropic hypogonadism or hypotha-
gery may not be necessary if the endocrine tests lamic–pituitary disease and hypogonadotropic
all point to anorchia. If there is evidence of tes- hypogonadism is the first step (Fig. 9.2). If the
ticular tissue by endocrine testing or by descent gonadotropins are high and there is no contribu-
after hCG stimulation or if the cryptorchidism is tory history such as surgery, radio or chemother-
unilateral, orchiopexy is indicated by age 1 year; apy, or autoimmune disease, consideration
further testicular damage will occur if the testes should be given toward a diagnosis that might be
are left in their warmer, intraabdominal location. confirmed by karyotype, such as Turner syn-
The findings of abnormal development of gen- drome or Klinefelter syndrome or their variants,
italia in boys, risk of low sperm count, and sus- even if the physical examination does clearly
ceptibility to testicular cancer as adults is suggest these possibilities.
postulated to be related to fetal exposure to toxins Differentiation between temporary constitu-
which are suspected to cause prenatal disruption tional delay in puberty and hypogonadotropic
of normal testis differentiation, the testicular dys- hypogonadism is difficult as both conditions have
genesis syndrome (TDS). low serum gonadotropin values. First consider-
A higher rate of hypospadias in Danish new- ation is always to rule out a CNS tumor or other
borns and smaller testes with lower serum inhibin serious conditions; complete neurologic evalua-
B compared with Finnish infants suggested that tion is performed and MRI considered if no other
environmental agents played a role in the differ- diagnosis or chronic disease is established. If
ence. This and a high frequency of reproductive midline abnormalities are present or anosmia is
problems among adult Danish men, including present, the diagnosis is likely to be hypogonado-
impaired semen quality, testicular cancer, and tropic hypogonadism. If a compelling family
increased rate of infantile testicular cancer in a history is found of delayed but ultimately sponta-
pattern that is attributed to environmental disrup- neous puberty (e.g., the father shaved later than
tors is described in the TDS. There may be a his peers or mother’s menarche was delayed) and
genetic component to susceptibility to the TDS there are no historical or physical findings of con-
based upon GWAS analysis indicating a gene X cern, the diagnosis is likely to be constitutional
environment basis for the disorder. delay in puberty. The presence of increased
serum gonadotropin values into the pubertal
Orchitis range on third-generation assays, a pubertal
Orchitis denotes an inflammation causing swell- response of LH after GnRH agonist, or increasing
ing of one or both of the testicles which may be a.m. serum testosterone values above 20 ng/dL
caused by an infection from many different types (a morning serum testosterone concentration
214 9 Disorders of Puberty

a Serum gonadotropins elevated


Yes Low

Karyotype Height and growth pattern

Abnormal Normal
Short for chronological Normal until Late-onset growth failure
age, but appropriate absence of
height for bone age growth spurt ? Signs of increased intracranial
pressure, ? Vision abnormalities,
Sense of smell ? Diabetes insipidus
Anosmia or No Yes
Normal hyposmia
Cranial MRI
Abnormal
Normal

47XXY-Klinefelter Primary testicular Constitutional Isolated Kallmann Consider CNS tumor


syndrome, 45XO- failure delay in growth gonadotropin syndrome malnutrition, chronic
Turner syndrome, Primary ovarian and puberty deficiency disease, Continued
Other failure hypothyroidism, surveillance
celiac disease

b Karyotype

Normal 46XX

Anatomic defect
of genital tract

Yes No
46XY 45XO or variant
Cranial MRI

Abnormal Normal

Syndrome of 45XO-Turner syndrome, Define defect, e.g., Cerebral Functional abnormality of


androgen or other variant (rare imperforate hymen neoplasm pulsatile LH release
resistance presentation in view of
normal secondary sexual
development, however)

Fig. 9.3 (a) Evaluation of absence of secondary sexual Cuttler L. Normal pubertal development. In: Rudolph CD,
development among boys at 14 years and girls at 13 years. Rudolph AM, eds. Rudolph’s Pediatrics. New York:
(b) Evaluation of primary amenorrhea with normal sec- McGraw-Hill, 2002:2093–2105, with permission)
ondary sexual development (revised from Styne DM,

over 20 ng/dL (0.7 mmol/L) indicates the likeli- reported at the usual age of puberty in hypogonad-
hood of pubertal development within 12 months) otropic hypogonadism, while a delay occurs in the
or an inhibin B value over 111 pg/mL (in an alter- increase in adrenal androgens along with a delay
native assay and study, the cutoff would be over in the appearance of all signs of secondary sexual
35 pg/mL) suggest that secondary sexual devel- development in constitutional delay in puberty.
opment will occur. A rise in nighttime peaks of Unfortunately, watchful waiting remains the main
gonadotropins is helpful but cumbersome to method of diagnosing the etiology of delayed
obtain. puberty: if a patient has not gone through the
Other strategies to differentiate CDG from HH changes of puberty spontaneously by age 18–19
have been attempted: for example, in some reports years, it is unlikely that he or she will do so.
an increase in adrenal androgens and the appear- The differential diagnosis of primary amenor-
ance of pubic hair without signs of gonadarche are rhea with normal secondary sexual development
Abnormalities of Puberty 215

may encompass the realm of chromosome anom- complications if an overdose occurs. The gel
alies, anatomic defects, or endocrine defects must not touch other children in the house
noted in this chapter. Historical and physical fea- directly or by clothes or towels or they also may
tures may point the way to possible diagnosis; become virilized. No appropriate oral prepara-
Fig. 9.3 demonstrates the reasoning that may be tion of testosterone is available or safe, as oral
used, based on the results of a karyotype determi- methyl testosterone may cause liver damage.
nation after initial evaluation is complete. If no sign of spontaneous puberty occurs in the
3 months after the treatment ceases, another
Treatment of Delayed Puberty course of testosterone can be offered after the
While psychological effects are caused by interval. Patients with known hypogonadotropic
delayed puberty due to an immature appearance, hypogonadism will have to receive testosterone
short stature appears to affect subjects more. As a therapy continuously in increasing doses until
general rule, boys are more distressed than girls. the final dose of 200–400 mg/month is reached
Psychological status should be investigated and over a 6–12-month period. Lower doses may be
support offered if necessary. given every 2 weeks rather than larger doses
Sex steroid administration is used to promote every 3–4 weeks to minimize psychological
pubertal development in hypogonadotropic hypo- effects of the waxing and waning testosterone
gonadism and also is useful temporarily in concentrations as higher doses are reached.
selected cases of constitutional delay. If the dif- Some men may require hCG injections in addi-
ference between the two cannot be established, 3 tion to testosterone to promote pubic hair devel-
months of low-dose sex steroid therapy can be opment and to achieve a more normal adult
used in patients feeling the pressure of an imma- appearance. Investigation into the use of LH and
ture appearance who are not comforted by the FSH rather than androgen replacement for boys
thought of “waiting for nature to take its course.” is under investigation as a more physiologic
The goal is to cause some progression of second- manner of promoting maturation of the testes
ary sexual development without advancing bone than sex steroid replacement. Replacement with
age and decreasing adult height. DHEA is a method sometimes used to increase
Individuals with hypogonadotropic hypogo- pubic hair development.
nadism have a risk of decreased bone density as Patients with GH deficiency who have been
adults. Since most teenagers do not get recom- treated with GH regularly since an early age usu-
mended doses of calcium or vitamin D, encour- ally can receive the sex steroid treatment regimen
aging adequate replacement is always wise. described earlier. If they remain quite short or
While genetics affects adult bone density signifi- have not been treated long before the teenage
cantly, administration of calcium and vitamin D years, testosterone therapy may be withheld for a
according to daily recommendation and sex ste- while or started at a lower dosage to ensure that
roids when appropriate may allow a closer maximal growth is reached before epiphyseal
approximation of the normal curve of bone den- fusion eliminates the ability to respond to GH.
sity with age. While investigators have used GnRH analogue to
After age 14 years (the upper age at onset of delay the progression or onset of puberty to allow
normal pubertal development), boys with HH increased growth on GH as an experimental
may be given 50 mg of testosterone enanthate therapy, efficacy is not supported.
intramuscularly every month for 3 months. Aromatase inhibitors inhibit the conversion of
Testosterone may be administered by skin androgens to estrogens, and since it is estrogen
patches or by the daily use of gel (however nei- that advances bone age and ultimately causes ces-
ther approved by the FDA for those under 16 sation of growth, this was found to increase adult
years) if the patient wishes to avoid injections. height. Thus, letrozole and anastrozole have been
Since testosterone enanthate lasts for a month, successfully used in boys with constitutional
daily treatment lowers the risk of long-term delay to allow achievement of a taller adult height.
216 9 Disorders of Puberty

Testes may become much larger than typical and with a family history of venous embolism may
plasma testosterone rises substantially during be at increased risk by estrogen therapy; testing
therapy. Side effects are reportedly rare but may for factor V Leiden may be performed (this is
include abnormalities in the shape of vertebrae. not a standard recommendation for all subjects
Theoretically adult bone density could be with out such a history at this time). Similar to the
decreased with the use of aromatase inhibitors, case in boys with respect to testosterone therapy,
but there is no proof of this concern at present. girls with GH deficiency should not receive long-
This is not an FDA-approved therapy and must be term or higher-dose estrogen therapy until growth
considered experimental and is only used in con- rate is normalized by GH administration.
trolled studies at present. A progestational agent such as medroxypro-
Girls with delayed puberty may be treated gesterone acetate (5 mg) is given on days 12–21
with low-dose estrogen therapy at 13 years if of the cycle after breakthrough bleeding begins to
CDP is suspected or earlier for HH or ovarian achieve a more normal endocrine milieu and to
failure. While oral therapy with ethinyl estradiol decrease the risk of uterine neoplasia from the
or conjugated estrogens has been the classic estrogen itself.
approach, there is concern about the nonphysio- Patients with Turner syndrome (the syndrome
logical manner of administration and the meta- of gonadal dysgenesis) can benefit from the treat-
bolic effects of oral estrogen first passing through ment of their growth deficiency. Growth rate can
the liver before reaching the true target tissue of be increased in Turner syndrome with adminis-
the breast and uterus. If oral therapy is to be used, tration of GH with a resulting increase in adult
17-beta estrogen is considered more physiologic height. The addition of oxandrolone, a weak
than ethinyl estradiol. Topical estrogen therapy is oral androgen, is reported to increase the effect
now preferred as it may carry less risk than oral of GH on growth. Side effects of oxandrolone
estrogen therapy of hypertension, gallstones, include pubic hair development, enlargement of
increased fat mass, decreased insulin sensitivity, the clitoris, and lowering of the voice, so this
and increased triglycerides (topical estrogen does agent is added only after the age of onset of nor-
not, however, increase HDL cholesterol and mal puberty, if at all. Previously girls with Turner
decrease low-density lipoprotein [LDL] cholesterol, syndrome were not given estrogens until the late
as does oral estrogen). Estrogen-impregnated teenage years for fear of decreasing their adult
patches are available for topical treatment but are height by advancement of their bone age; now,
not FDA approved in children. 17-Beta estradiol low-dose estrogens are started earlier to allow
patches (0.025 mg/patch; Vivelle Dot matrix secondary sexual development at an appropriate
patch) may be cut into fragments the size of age, such as 13 years, and to reduce psychoso-
one-eighth to one-fourth of a patch fragment to cial pressure and some studies suggest that it can
initiate pubertal development with more physio- be started even earlier. The combination of GH
logic estrogen values. After several months, and estrogen has not increased adult height more
and after the appearance of breakthrough bleed- than GH alone in studies when the estrogen is
ing, estrogen is given only on days 1–21 of the started early.
month. Withdrawal bleeding should then occur The infertility of Turner syndrome has also
regularly each month after the end of the estrogen been addressed. With the appropriate endocrine
administration. support, a donated ovum may be fertilized, by
Because of the concern over an increase in using the technique of in vitro fertilization, and
uterine carcinoma with exogenous estrogen transferred to a subject with Turner syndrome,
treatment, it is recommended that a girl taking but this method has great risk. Affected individu-
estrogens have a pelvic examination yearly. Girls als may be able to maintain a pregnancy with
and parents must be warned about increased risk of appropriate endocrine support to term and give
breast cancer with estrogen therapy, especially in birth. There is risk of uterine rupture in this situ-
those with a family history of this disease. Those ation, and there a 2 % mortality rate is reported,
Abnormalities of Puberty 217

so this course is not a standard of care. Of course, Table 9.4 (continued)


adoption is a well-recognized option for any Cranial irradiation
individual who wishes to be a parent whether True precocious puberty after late therapy for
infertile or not. congenital virilizing adrenal hyperplasia or other
previous chronic exposure to sex steroids
Peripheral (or incomplete) isosexual precocity
(hypothalamic GnRH independent)
Sexual Precocity (Table 9.4) Isosexual precocity
Boys
Sexual precocity in a boy is the development of Gonadotropin-secreting tumors
secondary sexual characteristics before age hCG-secreting CNS tumor (e.g.,
9 years. In the past the accepted onset of preco- chorioepithelioma, germinoma, teratoma)
cious puberty was before 8 years of age in girls, hCG-secreting tumors outside the CNS
but the obesity epidemic and other implicated fac- (hepatoma, teratoma, choriocarcinoma)
tors convinced most authorities that the age should Increased androgen secretion by adrenal gland or
be set lower. The age of diagnosis remains contro- testis
Congenital adrenal hyperplasia (CYP21 and
versial in girls, but the Pediatric Endocrine
CYP11B1 deficiencies)
Society presently states that when a girl has signs
Virilizing adrenal neoplasm
of puberty before her seventh birthday if Leydig cell adenoma
Caucasian or sixth if African American, the girl Familial testotoxicosis (sex-limited autosomal
has precocious puberty. Further analysis sug- dominant pituitary gonadotropin-independent
gested that these early limits of normal puberty precocious Leydig cell and germ cell maturation)
are best applied to girls who are overweight or Cortisol resistance syndrome
Girls
Table 9.4 Causes and classification of sexual precocity Ovarian cyst
Estrogen-secreting ovarian or adrenal neoplasm
Central (or complete) isosexual precocious puberty
Peutz–Jeghers syndrome
(GnRH-dependent sexual precocity or premature
activation of the hypothalamic pulse generator) Severe primary hypothyroidism
Familial or constitutional central precocious puberty Both sexes
Idiopathic true precocious puberty McCune–Albright syndrome
CNS tumors Iatrogenic or exogenous sexual precocity
(including unintentional exposure to estrogens in
Hamartoma of the tuber cinereum
food, drugs, or cosmetics or to topical androgens
Optic glioma associated with neurofibromatosis used by male members of the household)
type 1 or isolated
Contrasexual precocity
Hypothalamic astrocytoma
Boys
Craniopharyngioma
Estrogen-secreting adrenal or testicular tumor
Ependymoma
Exposure to exogenous estrogen
Other CNS disorders
Girls
Encephalitis
Androgen-secreting adrenal or ovarian tumor
Static encephalopathy
Late-onset CAH
Brain abscess
Exogenous androgen exposure (e.g., due to
Sarcoid or tubercular granuloma fathers’ improper use of topical testosterone)
Head trauma Variations of pubertal development
CNS surgery Premature thelarche
Hydrocephalus Premature isolated menarche
Arachnoid cyst Premature adrenarche
Myelomeningocele Adolescent gynecomastia of boys
Vascular lesion
CNS central nervous system, hCG human chorionic
(continued) gonadotropin, GnRH gonadotropin-releasing hormone
218 9 Disorders of Puberty

obese as normal pubertal development is rarer lishing the diagnosis although careful genetic
at those ages in girls within the normal range of evaluation of family patterns suggests that the
weight. However, any girl with the onset of occurrence of early puberty is often not reported
puberty before 8 years must be considered with a accurately. While slightly early puberty might be
high index of suspicion for a disease as the etiol- the mirror image of constitutional delay in
ogy so as not to miss subtle findings of a condition puberty, in which there is a family tendency in
such as a brain tumor. A careful search for historical normal subjects toward delayed onset of puberty,
or physical features of organic disease must occur in either case a deviation from the normal limits
before a girl with puberty earlier than 8 years is of age of onset of puberty is not to be taken
considered normal. The child must have abso- lightly and must be considered a disorder until
lutely no sign of central nervous system disorder adequate evidence proves it to be a normal varia-
or other conditions that might trigger pathologic tion. To consider a child who starts pubertal
precocious puberty. A girl who proceeds rapidly development slightly below the normal age
through puberty, who has menarche before age 9 guidelines as having a constitutional tendency,
years, or has neurological signs or other findings no signs of neurologic or other serious diseases
of disease accompanying such early puberty must must be present, and the sequence of events and
not be considered normal before a complete eval- progression must be the same as in normal
uation is completed. Thus, in any case in which puberty. With the recent finding of a familial
concern arises, investigation for a disorder that nature of CPP in more subjects, these borderline
might be causing precocious puberty is war- cases may be manifestation of these genetic
ranted, rather than simply relying on the age cri- variants.
teria stated earlier.
If the etiology of early puberty is the premature Idiopathic Central Precocious Puberty
maturation of the hypothalamic–pituitary axis, the If no tumor or other definitive diagnosis is found,
condition is denoted as central, complete, or true and if no family tendency exists, the diagnosis of
precocious puberty (CPP), which is a pituitary exclusion is idiopathic CPP. These patients mani-
gonadotropin-dependent condition; if the etiol- fest all of the endocrine findings of normal
ogy is autonomous secretion of sex steroids in puberty, albeit at an earlier age. Their progress
boys or girls or autonomous secretion of hCG in may be normal and continuous or slow and wax-
boys, the condition is peripheral precocious ing and waning. Boys with this condition will first
puberty (or incomplete precocious puberty or demonstrate symmetrical and progressive testicu-
pseudo-precocious puberty), a pituitary lar enlargement, just as found in normal puberty.
gonadotropin-independent condition. If the puber- Girls are brought to evaluation for idiopathic pre-
tal development is appropriate for sex but early it is cocious puberty more often than boys in the USA,
isosexual precocity and if virilization occurs in partially due to differing parental concerns about
girls or feminization in boys, it is contrasexual the social effects on girls of early pubertal devel-
precocity. Patients of all categories will have rapid opment compared with boys.
growth and skeletal maturation and, without treat-
ment, may fulfill the paradox of the tall child who Gain-of-Function Mutations
ceases growing early because of premature epiphy- in the Kisspeptin Axis
seal fusion and becomes a short adult. Inactivating mutations in the KISS1R cause
hypogonadotropic hypogonadism, but an auto-
Central (Complete or True) Precocious somal dominant KISS1R mutation is associated
Puberty (Fig. 9.4a) with CPP due to prolonged action of the
The onset of puberty occurring close to, but receptor compared to wild type (#176400
below, the lower age limits of normal pubertal PRECOCIOUS PUBERTY, CENTRAL).
development, without evidence of an organic dis- Likewise mutation in the KISS gene which
order, may possibly be a normal variant. A family codes for kisspeptin has been found in three
tendency toward this situation will assist in estab- children with precocious puberty. One of the
Abnormalities of Puberty 219

a Third ventricle Hypothalamus

Neuroendocrine
cell nuclei

GnRH secreting cells

Superior
hypophyseal Stalk
artery

Long portal
vessels Inferior
hypophyseal
artery
LH and FSH

Secreting cells

Anterior Posterior
pituitary pituitary

Hormone Short portal


secretion vessel

LH and FSH

Testosterone Estrogen
and inhibin and inhibin

Testes Ovary

Fig. 9.4 (a) Central precocious puberty in which (b) Peripheral precocious puberty in which the gonads
increased GnRH stimulates the anterior pituitary gland to autonomously secrete their respective sex steroids to
increase secretion of LH and FSH which stimulate the cause secondary sexual development. In this case the
gonads. While sex steroid secretion increases and causes increased sex steroids do suppress GnRH secretion, but
the physical changes of puberty, the steroids are unable to since GnRH is not involved in the etiology of this condi-
exert feedback inhibition to decrease the GnRH secretion. tion, puberty progresses

mutations appears to lead to prolonged survival makorin ring finger protein 3, is an important
and activity of kisspeptin from degradation. inhibitor of pubertal development during the
juvenile pause (#615346 PRECOCIOUS
MKRN3 PUBERTY, CENTRAL, 2; CPPB2). All of the
Recently loss-of-function mutations of MKRN3, children, mostly girls, inherited their mutation
an imprinted gene located on the long arm of from their fathers as the maternal allele of this
chromosome 15q11.2, in children with CPP led to gene is silenced by methylation, a case of monoal-
the consideration that this gene and its product, lelic expression. The familial nature of the condi-
220 9 Disorders of Puberty

b Third ventricle Hypothalamus

Neuroendocrine
cell nuclei

GnRH secreting cells

Superior
hypophyseal Stalk
artery

Long portal
vessels Inferior
hypophyseal
artery
LH and FSH

Secreting cells

Anterior Posterior
pituitary pituitary

Hormone Short portal


secretion vessel

LH and FSH

Testosterone Estrogen
and inhibin and inhibin

Testes Ovary

Fig. 9.4 (continued)

tion was not always apparent until careful cases of CPP, a CNS tumor must be considered
determination of family history was taken. Some first as the etiology, especially in boys; idiopathic
affected girls began puberty between 6 and 9 conditions are diagnoses of exclusion.
years of age providing another reason that CPP Hamartomas of the tuber cinereum are con-
below 8 years in girls may not be idiopathic. genital conditions composed of ectopic hypotha-
Treatment with GnRH agonist appears effective lamic tissue and usually contain GnRH in their
in most cases. neurons. They function as a supplemental hypo-
thalamus that operates outside of the normal
Central Nervous System Disorders inhibitory effects of the CNS on GnRH secretion.
CPP occurs more often in girls than in boys. They have characteristic locations and appear-
However, CNS tumors are found more often in ances on MRI, so biopsy is rarely necessary.
boys with precocious puberty than in girls. In all They are not neoplasms and do not enlarge.
Abnormalities of Puberty 221

Because of their sensitive location, hamartomas hyperplasia (CAH), after the removal of an adre-
of the tuber cinereum are rarely amenable to sur- nal androgen-secreting tumor, and after androgen
gical removal: medical therapy with GnRH ago- therapy for various disorders, such as anemia.
nists is the treatment of choice for this form of
precocious puberty. However, because intracta- Peripheral Precocious Puberty
ble gelastic (laughing) or other types of epilepsy and Exogenous Sex Steroids (Fig. 9.4b)
may accompany the hamartoma, surgical treat- Unregulated sex steroid secretion is the major
ment may be directed to the epilepsy but is rarely cause of peripheral precocious puberty in girls
used solely for precocious puberty. and in boys, although boys with tumors secreting
Ominous CNS tumors include germinomas hCG will also have peripheral precocious puberty
often located in the pineal gland. Germinomas as the hCG stimulates the testes. The only sites
may secrete hCG and cause incomplete preco- that can secrete sex steroids in an unregulated
cious puberty in boys (see later) or may activate manner in disease are the adrenal glands and the
the entire hypothalamic–pituitary axis and cause gonads.
central precocious puberty. Germinomas are
radiation sensitive. Other CNS tumors causing Girls
precocious puberty (or delayed puberty depend- Peripheral isosexual precocious puberty may
ing upon the location) are astrocytomas and develop in girls because of ovarian or adrenal
ependymomas; gliomas of the optic nerve or secretion of, or ingestion of or exposure to, estro-
hypothalamus may be isolated or part of neurofi- gen. Serum gonadotropins are suppressed, whereas
bromatosis. See Chaps. 3 and 5 for more discus- serum estradiol concentrations are elevated.
sion of these CNS tumors that also affect growth. Ovarian follicular cysts can secrete enough
Delayed rather than precocious puberty may estrogen to cause breast development, and when
be the outcome of tumors or injuries of the CNS, the cyst resolves, withdrawal bleeding can occur.
depending upon the location in the hypothala- Usually cysts are small and limited in effect, but
mus. Posterior hypothalamic locations of central some can secrete enough estrogen to cause serum
nervous system are the ones most likely to cause estradiol concentrations to increase as high as
precocious puberty. those found in tumors. Cysts may occur only
Other CNS causes of true precocious puberty once, but recurrent cysts are possible. Surgical
include space-occupying lesions or etiologies of intervention is rarely indicated for simple cysts.
increased intracranial pressure, including granu- Estrogen-secreting tumors include granulosa
lomas, suprasellar arachnoid cysts, hydrocepha- cell tumors (the most common), gonadoblasto-
lus, or head trauma including birth trauma. mas (which can arise in streak gonads of gonadal
Precocious puberty may be associated with cere- dysgenesis), lipoid tumors, and ovarian carcino-
bral palsy and other intellectual defects. mas. The majority of granulosa cell tumors can
be palpated by bimanual examination.
Other Causes of CPP Exogenous estrogen exposure may occur
Severe hypothyroidism can cause delayed through diet (chicken necks, beef, or veal have
puberty or, paradoxically, precocious puberty: contained estrogen because of improper com-
the latter, known as the Van Wyck–Grumbach mercial estrogen treatment to enhance growth
syndrome, appears due to cross-reaction with the of the animals), by ingestion of medications
elevated TSH concentrations interacting with the that may include estrogen (perhaps given to
FSH receptors. Any virilizing condition, when mother or grandmother), or even by skin
resolved, can trigger true precocious puberty, absorption after contact with estrogen-contain-
presumably because of maturation of the hypo- ing cosmetics or hair cream or lotions (laven-
thalamic–pituitary axis. This may occur after glu- der or tea tree oil has been implicated after
cocorticoid treatment is initiated for dermal application and some hair pomades
long-untreated virilizing congenital adrenal contain sex steroids).
222 9 Disorders of Puberty

Boys develops following successful treatment of the ear-


Boys can develop incomplete isosexual preco- lier peripheral PP, GnRH agonist is then effective
cious puberty either by autonomous secretion or in the CPP phase.
ingestion of sex steroids or by tumor production If hCG stimulates the testes to produce testos-
of hCG, which will cause testosterone secretion terone, the testes will be slightly enlarged (more
from Leydig cells due to hCG interacting with than 2.5 cm) but will not be as large as found in
the LH receptors. hCG production causes no true precocious puberty, because the seminifer-
endocrine or physical effects in girls. Serum LH ous tubules, which are normally responsive to
and the LH response to GnRH will be sup- FSH, will not increase to pubertal size since no
pressed because of autonomous testosterone increased FSH secretion occurs. hCG-secreting
secretion in peripheral gonadotropin-independent tumors include hepatomas, hepatoblastomas,
precocious puberty. teratomas or chorioepitheliomas of the gonads,
Androgen secretion may occur due to mediastinum, and retroperitoneum, as well as
21-hydroxylase or 11-hydroxylase deficiency germinomas of the pineal gland. A hepatoblas-
CAH, adrenal carcinomas, and interstitial cell toma may quickly prove to be fatal after the
tumors of the testes. A Leydig cell tumor is seen diagnosis of precocious puberty is made.
with irregular enlargement of one or both testes. The McCune–Albright syndrome (#174800
With adrenal hyperplasia, the testes will usually be MCCUNE–ALBRIGHT SYNDROME; MAS at
of prepubertal size in younger boys since the 20q13.2) involves the triad of café au lait spots,
androgen emanates from the adrenal glands. fibrous dysplasia of the long bones (cysts are nota-
However, a testicular adrenal rest tumor composed ble on radiograph or bone scan, and pathologic
of ACTH-responsive tissue located in the testes, a fractures are found), and either central or incom-
common but ectopic location for adrenal tissue plete precocious puberty. Other forms of increased
originating in fetal life, may enlarge later in endocrine activity such as Cushing syndrome,
CAH; this is denoted as testicular adrenal rest tis- hyperthyroidism, pituitary gigantism, and hypo-
sue, or TART (see Chap. 6). phosphatemic rickets are found in McCune–
Premature Leydig and germinal cell maturation Albright syndrome. The wide-ranging effects of
or male limited precocious puberty (#176410 this condition occur because of somatic mutations
PRECOCIOUS PUBERTY, MALE-LIMITED at in the Gα-stimulatory protein of the adenyl cyclase
2p21) is a rare condition but is instructive in the pathway. Thus the LH receptor and other mem-
understanding of puberty, as it results from a unique brane-based protein hormone receptors are con-
error of biology. A mutation in the seven-trans- stitutively activated by the Gα-stimulatory
membrane domain of the LH receptor causes the protein and always functioning, even in the
LH receptor to be constitutively activated so that absence of the stimulatory ligand. Multiple
testosterone is produced even in the absence of tissues (e.g., skin, bone, Leydig cells, follicular
LH. Affected boys demonstrate androgen effects at cells, thyroid cells, adrenal cortical cells, and
an early age, and often a family history of affected pituitary cells) are activated in a random distribu-
fathers or uncles is found in this male limited auto- tion in the body due to somatic mutations.
somal dominant disorder. Remarkably, at the time
of normal puberty, these boys switch from periph- Contrasexual Precocity: Feminization
eral gonadotropin-independent precocious puberty in Boys and Virilization in Girls
to normal GnRH-dependent pubertal physiology
that ultimately can lead to fertility; indeed central Boys
precocious puberty can follow suppression of the Feminization in a boy before the age of puberty
autonomous phase of testosterone secretion. is rare. Rarely, an estrogen-secreting adrenal
Treatment may be effective with an antiandrogen adenoma or a chorionepithelioma causes gyne-
such as flutamide or nialamide and an aromatase comastia. The treatment of adenomas or carci-
inhibitor such as letrozole in decreasing the effects noma of the ovary, testis, or adrenal gland is
of androgen secretion early in life. When CPP surgical correction.
Abnormalities of Puberty 223

Aromatase Excess Syndrome (17-hydroxypregnenolone), DHEA, and DHEAS,


Gynecomastia in prepubertal boys can be caused as well as decreased secretion of aldosterone and
by increased extraglandular aromatization of cortisol in the severe form. Severely affected
C19 steroids of adrenal origin, such as andro- patients have mineralocorticoid and glucocorti-
stenedione, and subsequent increased extraglan- coid deficiencies and may die in infancy. Excess
dular estrogen production in sporadic or familial adrenal androgens lead to virilization in utero and
cases. The autosomal dominant form leads to to ambiguous external genitalia, including clitoral
excess estrogen synthesis from C19 precursors enlargement in females with continued viriliza-
due to aromatase overexpression, especially in fat tion after birth. Milder forms of this disorder can
and skin; it is a consequence of gain-of-function cause hirsutism in women. Women with a 46,XY
mutations of CYP19, the gene that encodes aro- phenotype and incomplete forms of androgen
matase, resulting from a chromosome arrange- resistance syndrome or 17β-HSD type 3 defi-
ment that gives rise to a cryptic promoter. ciency may have virilization as well as breast
development at the time of expected puberty.
Feminizing Testicular Tumors Aromatase deficiency due to mutations in the
Feminizing testicular tumors may cause gyneco- CYP19 gene, which encodes aromatase, is associ-
mastia in boys younger than 6 years of age who ated with intrauterine masculinization of the
have the Peutz–Jeghers syndrome. Aromatase is external genitalia in affected 46,XX individuals
absent or is present in barely detectable amounts in and also with progressive virilization, lack of
prepubertal testes, but maximal amounts appear in female secondary sex characteristics, multicystic
late puberty. In normal testes, aromatase is pre- ovaries at the age of puberty, tall stature, and
dominantly present in the Leydig cells, but in tes- osteopenia.
ticular tumors of Sertoli cells or Leydig cells Cushing syndrome resulting from adrenal car-
(e.g., associated with the Peutz–Jeghers syn- cinoma usually manifests as growth failure with
drome), the Sertoli cells of the tumor express aro- or without virilization, obesity, and moon facies;
matase. Both testes may be enlarged, and the striae may not appear until months to years later.
histology indicates sex cord or Sertoli cell tumors
that form annular tubules and often have areas of Syndrome of Glucocorticoid Resistance
calcification; increased estradiol secretion is The syndrome of glucocorticoid resistance has
noted in the basal state, and a further rise occurs variable manifestations. Some patients demon-
after hCG administration. Sonography or MRI of strate hyperandrogenic signs such as acne, hir-
the testes may be useful in making the diagnosis. sutism, male-type baldness, menstrual
Prepubertal gynecomastia can also be caused irregularities, and oligoanovulation and infertil-
by neurofibromatosis. ity. Dexamethasone decreases the excessive
adrenal androgen secretion, virilization, and
Girls advancing bone age found in general glucocorti-
coid resistance.
Adrenal Causes of Virilization
Virilization in a girl indicates serious organic dis- Virilizing Ovarian Tumors
ease except for premature adrenarche which leads Arrhenoblastoma, also called Sertoli tumor of the
to only modest androgen effects. CAH resulting ovary, is the most common virilizing ovarian
from 21-hydroxylase or 11β-hydroxylase defi- tumor, but it is rare in children. Somatic missense
ciency or from androgen-producing tumors of the mutations affecting the RNase IIIb domain of
adrenal can cause virilization. Nonclassic or late- DICER1 that alter DICER1 function in a manner
onset forms of CAH do not demonstrate ambigu- that perturbation of microRNA processing may
ous genitalia, but there is evidence of androgen be oncogenic are common in these nonepithelial
effect in prepuberty or the teenage years. 3β-HSD/ ovarian tumors. Lipoid-cell tumors of the ovary
Δ4,5-isomerase deficiency is a rare type of CAH and gonadoblastomas of the ovary are even more
characterized by elevated levels of Δ5-17P unusual sources of androgens.
224 9 Disorders of Puberty

Endocrine Disruption in Puberty it is important to continue to observe children


with premature thelarche over time. Exaggerated
There are rising concerns that endocrine disrupting thelarche is found in girls with premature thelar-
chemicals (EDCs), defined as “an exogenous che with some advancement of the bone age and
substance that causes adverse health effects in an increase in growth rate.
intact organism, or its progeny, secondary to
changes in endocrine function,” exert many Premature Adrenarche
effects on growth and development of children Premature adrenarche is a benign self-limited
including pubertal development. Far more con- appearance of a small amount of pubic hair, com-
clusive evidence of an adverse effect of EDCs edones, axillary hair, or odor, which usually
comes from animal rather than human studies, occurs after age 6 years. The normal increase in
and some of the data on human beings derives adrenal androgens, including DHEA and its sul-
from industrial accidents and very-high-level fate (DHEAS), occurs earlier in this condition
exposure rather than the lower level exposures than in an unaffected child, and the pubic hair
most individuals experience, so EDCs must be then follows the increase in DHEAS. Thus a
studied in more detail in human beings. 6–8-year-old with Tanner stage 2–3 pubic hair
Nonetheless, there is a significant concern that may have a DHEAS value characteristic of a
the exposure of the fetus or developing child to 12–13-year-old. The rest of pubertal develop-
the multitude of industrial chemicals in the envi- ment, such as testicular development in a boy or
ronment either delay or advance pubertal devel- breast development in a girl, will occur at a nor-
opment. Parents will often ask about these issues mal age. A slight increase in growth rate may be
Legislatures have acted in some states to try to noted, along with a slight advancement of bone
decrease chemical exposures. University centers age, but neither should be pronounced, or a more
are studying this issue and developing guidelines serious virilizing disorder is likely.
to help parents avoid toxic exposures. Late-onset CAH may have an initial appear-
ance indistinguishable from that of premature
adrenarche. Thus clinical follow-up is indicated,
Variations of Early Pubertal even if it appears that the child has the benign
Development condition, premature adrenarche. If any doubt
exists that the condition seems to be more severe
Premature Thelarche than premature adrenarche, or if a family history
Premature thelarche is a benign condition of of the same condition is found, testing for CAH is
unilateral or bilateral breast development, usu- indicated by using basal adrenal metabolite eval-
ally in a girl younger than 3 years. Minimal or uation as well as measurement after ACTH stim-
no other signs of estrogen effect are present (i.e., ulation (see Chaps. 10 and 15). Polycystic ovarian
no dulling of the vaginal mucosa, little areolar syndrome (PCOS) also appears in some cases
(nipple) development or pigmentation, and no similar to premature adrenarche. A history of
increase in growth rate). Serum estradiol values SGA birth may precede the onset of premature
are prepubertal, as the follicular cyst thought to adrenarche. (PCOS is discussed in Chap. 13)
cause this condition is often spontaneously An adrenal tumor or other more serious form
resorbed by the time the patient comes to evalu- of virilization must manifest limited virilization in
ation. Some evidence exists that FSH is higher its earlier stages and may be mistaken for
in premature thelarche than in age-matched chil- premature adrenarche, so follow-up of premature
dren without thelarche, but the serum LH and adrenarche, although usually benign, is indicated.
the LH response to GnRH are prepubertal. Some
children with central precocious puberty may at Gynecomastia
the beginning of this condition appearing very Gynecomastia is the appearance of breast devel-
much as if they have premature thelarche. Thus, opment in a male. Physiologic or pubertal gyne-
Endocrine Disruption in Puberty 225

comastia is a variation of normal that is seen in a or signs of puberty. Testicular adrenal rests (TART)
boy who, usually, has reached Tanner stage 2–3 in CAH can cause bilateral macroorchidism as can
pubertal development. In the majority of cases, lymphoma. Macroorchidism is a feature of severe
the condition disappears within 1–3 years, but in aromatase deficiency in young male adults and in
some cases, it may last longer and require surgi- men with an FSH-secreting pituitary macroade-
cal removal. The longer the tissue remains, the noma. Bilateral megalotestis (testicular volume,
more likely it will not spontaneously resolve, 26 mL) in adults can occur as a normal variant.
because scar tissue ultimately develops in the Testes will enlarge during treatment with aroma-
breast tissue. Obese subjects may have a reduc- tase inhibitors.
tion in this glandular tissue or its prominence by The fragile X syndrome is associated with
reducing their weight. Thinner boys with several developmental delay, a long face and large prom-
years of gynecomastia may require surgery to inent ears, and macroorchidism in 80 % of
resolve the condition. affected pubertal boys. The enlarged testes are
Pathologic gynecomastia will occur in caused by increased interstitial volume and
Klinefelter syndrome; breast cancer may develop excessive connective tissue, including increased
in the tissue affected by gynecomastia in peritubular collagen fibers, rather than by an
Klinefelter syndrome. Gynecomastia is also a increase in the seminiferous tubules. Enlargement
feature of Reifenstein syndrome (#312300 of the testes is demonstrable in the prepubertal
REIFENSTEIN SYNDROME), or any form of period in most patients with fragile X syndrome,
partial androgen resistance, as well as in rare pre- but the onset of true macroorchidism (>4 cm)
pubertal boys with 11β-hydroxylase deficiency occurs only in the later prepubertal period.
(*202010 ADRENAL HYPERPLASIA,
CONGENITAL, DUE TO 11β-HYDROXYLASE
DEFICIENCY at 8q21) or aromatase excess syn- Differential Diagnosis of Precocious
drome (139300 GYNECOMASTIA, Puberty
HEREDITARY). Medical therapy has been tried
without success in gynecomastia, so surgery is A girl with early-onset breast development may
the only present treatment for persistent gyneco- have premature thelarche (usually with more sub-
mastia. Prepubertal gynecomastia indicates tle breast development) or estrogen secretion due
exposure to or pathologic secretion of estrogen to peripheral precocious puberty or gonadotropin-
and must be carefully investigated. stimulated estrogen secretion due to central preco-
cious puberty if thelarche is more substantial.
However, if a girl has early breast development and
Macroorchidism an appropriate stage of pubic hair development for
the degree of breast development, she probably has
Macroorchidism is defined as testes twice the CPP, as no other condition is likely to produce both
normal size for age without androgenization. It is estrogen and androgen in balanced amounts
a rare manifestation of the McCune–Albright (Fig. 9.4a). A boy who shows signs of virilization
syndrome and an occasional finding in prepuber- with symmetrically enlarging testes of homoge-
tal boys with long-standing primary hypothy- neous appearance and consistency to palpation is
roidism. In the McCune–Albright syndrome, an likely to have CPP: bilateral testicular tumors are
activating mutation in the Gsα gene primarily alternative etiologies of enlarging testes, but it is
expressed in the Sertoli cell can cause macroor- unlikely that they will be symmetrical and homo-
chidism due to Sertoli cell proliferation and geneous and have regular surfaces. Gonadotropin-
hyperfunction with increased concentration of independent Leydig and germ cell maturation
serum inhibin B and AMH; there may be no leads to a small degree of enlargement of testes
increase in testosterone levels due to absence of since the seminiferous tubules are not as affected
Leydig cell hyperplasia, elevated gonadotropins, as Leydig cells, but the testes do not reach full
226 9 Disorders of Puberty

pubertal size. If the testes are not enlarging even gonadotropin release. When GnRH-A sup-
though the patient exhibits androgen effects, an presses gonadotropin secretion, sex steroid
adrenal source of androgen products must be sus- secretion decreases, the rate of bone-age
pected, such as an adrenal tumor or adrenal enzyme advancement decreases, as does the rate of rapid
defect, or exogenous androgens as the etiology. growth, and the height prognosis of treated
If significant isosexual pubertal development is patients improves if treatment is started early,
noted (breast development in a girl or virilization usually before 6 years. The effects are revers-
in a boy), determination of sex steroid and serum ible, as those patients whose therapy is discon-
gonadotropin concentration (in modern, sensitive tinued increase gonadotropin secretion to
pediatric assays) is performed. Serum gonadotro- pubertal values, following the same pattern as is
pin concentrations and GnRH agonist-stimulated usually found in the early stages of normal
LH and FSH will reveal pubertal values in CPP, puberty, and fertility is reported in adult life.
but the LH values will be prepubertal in preco- GnRH-As are commercially approved for the
cious adrenarche or in precocious thelarche (see treatment of central precocious puberty in the
Fig. 9.5). If the diagnosis is CPP, a search for a form of monthly or three-monthly injections,
CNS abnormality is indicated, and an MRI is per- daily injections, and yearly surgical implants.
formed to determine whether a hamartoma of the GnRH agonist therapy is not necessary to ensure
tuber cinereum or an expanding neoplasm is pres- an adequate adult height in a patient who has
ent. Boys more often than girls will have a brain slowly progressive or waxing and waning preco-
tumor as the cause of their true precocious puberty cious puberty that commences at an age only
(Table 9.5). slightly younger than the normal age at the onset
If sex steroids are elevated but gonadotropins of puberty. Further, GnRH agonist treatment of
are suppressed, autonomous sex steroid secretion puberty with the onset after 6 years of age is less
is likely to be found. If beta hCG is elevated, an likely to increase adult height.
hCG-secreting tumor is suggested as modern Testolactone and letrozole are aromatase inhib-
beta hCG assays do not cross-react with LH. An itors that stop estradiol and estrone biosynthesis.
MRI is invoked to search for liver, CNS, medias- These added to antiandrogens such as flutamide or
tinum, or other locations for the hCG-secreting nilutamide are used in McCune–Albright syn-
tumor. Alternatively, exogenous sex steroids can drome in boys with peripheral precocious puberty
cause these laboratory findings. and in familial gonadotropin-independent Leydig
cell and germ cell maturation to decrease viriliza-
tion. Ketoconazole, an antifungal agent, causes a
Treatment of Precocious Puberty 17–20 lyase block in the testosterone biosynthetic
pathway and has been used to decrease testoster-
Gonadotropin-releasing hormone analogues one secretion in familial gonadotropin-
(GnRH-As) are extremely potent versions of independent Leydig cell and germ cell maturation;
GnRH which have an extended time of action. adrenal suppression is a side effect of such treat-
These agents cause downregulation of GnRH ment and adds to the risk of treatment.
receptors, such as would occur during a constant Peripheral precocious puberty can be followed
GnRH infusion, thereby decreasing episodic by true precocious puberty after treatment if the

Fig. 9.5 (continued) before 7 years among white girls African-American girls. DHEAS dehydroepiandrostene-
and 6 years among African-American girls (revised dione sulfate, 17α-OHP 17α-hydroxyprogesterone,
from Styne DM, Cuttler L. Normal pubertal develop- GnRHA gonadotropin-releasing hormone agonist
ment. In: Rudolph CD, Rudolph AM, eds. Rudolph’s (reprinted from Styne DM, Cuttler L. Normal pubertal
Pediatrics. New York: McGraw-Hill, 2002:2093–2105, development. In: Rudolph CD, Rudolph AM, eds.
with permission). (c) Diagnostic algorithm for pubic Rudolph’s Pediatrics. New York: McGraw-Hill,
hair before 7 years among white girls and 6 years among 2002:2093–2105, with permission)
Endocrine Disruption in Puberty 227

a Possibility of exogenous
androgens or hCG
Yes No

Testicular size

Longest diameter < 2.5 cm Longest diameter > 2.5 cm

Adrenal etiology Serum hCG elevated


Yes No
Asymmetrical
17OHP DHEAS Cortisol elevated, ACTH Serum LH suppressed enlargement
elevated elevated elevated, no signs of
Yes No (Basal LH or LH
Cushing syndrome
response to
GnRHA is pubertal)
Exogenous P450C21 Adrenal Glucocorticoid hCG Testotoxicosis or True precocious Testicular tumor or
androgen or deficiency virilizing resistance secreting other autonomous puberty adrenal rest with CAH
hCG exposure tumor or tumor testosterone
late-onset secreting lesion
3β HD of testes
deficiency

b Basal or GnRHA
stimulated levels of
LH are pubertal
Yes No

Central Serum estradiol


precocious puberty at pubertal levels
No Yes

Cranial MRI
Ultrasound of
uterus and ovary
Normal Abnormal Normal Abnormal

Cafe au lait spots

No Yes

Idiopathic Hamartoma of tuber Premature Consider: Estrogen-secreting Ovarian cyst McCune−Albright


precocious cinereum, CNS thelarche adrenal neoplasm, extrinsic or tumor syndrome
puberty tumor, or other lesion estrogen exposure

c
DHEAS above prepubertal levels
Yes No

Basal or ACTH-stimulated
17-OHP levels elevated
Yes No

Free testosterone
level increased
Yes No

Cortisol and ACTH


levels elevated
Yes No

Virilizing Late-onset P450021 Virilizing Cushing syndrome Premature


adrenal deficiency ovarian or glucocorticoid adrenarche
tumor neoplasm resistance

Fig. 9.5 (a) Diagnostic algorithm for precocious Rudolph CD, Rudolph AM, eds. Rudolph’s Pediatrics.
puberty among boys before age 9 years (revised from New York: McGraw-Hill, 2002:2093–2105, with per-
Styne DM, Cuttler L. Normal pubertal development. In: mission). (b) Diagnostic algorithm for breast development
228 9 Disorders of Puberty

Table 9.5 Differential diagnosis of precocious puberty


LH response to Serum sex
Serum gonadotropin GnRH or GnRH steroid
concentrations agonist concentrations Gonadal size Miscellaneous
Central (complete or true) precocious puberty
Pubertal Pubertal pattern Pubertal values Normal pubertal MRI scan of head to
enlargement of rule out a central
gonads in males nervous system tumor,
hamartoma, or other
central nervous system
lesion
Incomplete precocious puberty
Males
Chorionic gonadotropin- High basal hCG or High or Slight to moderate Hepatic tumor must be
secreting tumor, high LH that does not rise pubertal values enlargement of considered. MRI scan
hCG (or LH in cross- with GnRH, but gonads of head if hCG-
reactive assay), positive β-LH is low and secreting central
pregnancy test, but β-LH does not rise nervous system tumor
(specific assay) is low suspected. LH assay
may measure hCG, so
both results may be
high while β-hCG is
high and β-LH, in an
assay which does not
detect hCG, is low
Leydig cell tumor, Prepubertal or Extremely high Irregular asymmetric
prepubertal suppressed pattern testosterone enlargement of testes
(gonadotropins low)
Familial gonadotropin- Prepubertal or Pubertal or Testes longer than Found in sex-limited
prepubertal independent suppressed pattern higher values 2.5 cm but smaller dominant patterns
sexual precocity with than expected for
premature Leydig and stage of pubertal
germ cell maturation development
prepubertal (low)
Females
Granulosa cell tumor Prepubertal or Extremely high Ovarian enlargement Granulosa cell tumor
prepubertal (follicular suppressed pattern estradiol on physical, CT, is usually palpable on
cysts may have similar MRI, or sonographic rectal examination
presentation) examination
Follicular cyst Prepubertal or High to Cysts may be visible Withdrawal bleeding
prepubertal suppressed pattern of extremely high on sonogram may occur when
gonadotropins are low LH secretion. FSH estradiol estrogen levels
secretion may rise decrease. Cysts may
above normal range recur

condition has been uncontrolled for a long period Psychological support is helpful to both child
and the sex steroids have exerted a maturational and parents. Their larger size may make children
effect on the hypothalamic–pituitary axis. In this with precocious puberty the center of unwanted
situation GnRH-A might be effective even though attention. Children with true precocious puberty,
it was not effective during the peripheral preco- if intellectually and socially appropriate, may be
cious puberty portion of the condition. able to accept accelerated school placement so
Polycystic Ovarian Syndrome 229

that they can be placed in a class with children common in PCOS in the USA but less common in
whose size is closer to their own. Girls with pre- PCOS in other countries suggesting an environ-
mature menstrual periods should be prepared for mental influence on the development of obesity
the event before menarche and supported through related to PCOS. PCOS is found in familial pat-
this difficult time. Of further concern is the pos- terns and the unaffected sisters and even the broth-
sibility that the patient could be the target of sex- ers of the affected individuals have endocrine
ual abuse, with the additional element of potential manifestations of hyperandrogenism. A correlation
fertility. Boys with precocious puberty and high of low birth weight and the development of
testosterone values will be prone to aggressive polycystic ovarian syndrome has been reported.
activity and may masturbate publicly, but are Premature adrenarche is felt to be harbinger in
unlikely to seek out sexual activity in the absence some girls of the development of PCOS, but this is
of social maturation. In some cases, the treating not universal.
physician can help the family through this stress- There are various criteria for the diagnosis
ful time, but psychological counseling should be of polycystic ovarian syndrome, but they are
considered in the more significant cases. validated in adults in most cases. The Rotterdam
Early menarche is associated with a higher criteria for polycystic ovarian syndrome are in
risk of breast cancer; thus, affected girls should common use and include evaluation for the
be counseled as adults about screening for this presence of polycystic ovarian appearance on
disease. ultrasound examination. This consideration is
A discussion of transgender youth is beyond more difficult to establish in teenagers as a
the scope of this volume, but it should be men- transvaginal ultrasound may not be appropriate
tioned that delaying puberty in these patients and the obesity often found in polycystic ovar-
while psychological counseling and determina- ian syndrome decreases the accuracy of a trans-
tion of the ultimate course of therapy is proceed- abdominal ultrasound. The Endocrine Society
ing may be accomplished using GnRH agonists has provided guidelines for the diagnosis of
(see Chap. 8). polycystic ovarian syndrome in teenage girls.
According to the guidelines, diagnosis of PCOS
in an adolescent girl is made if there is clinical
Polycystic Ovarian Syndrome and/or biochemical evidence of hyperandrogen-
ism (after exclusion of other pathologies) in the
Polycystic ovarian syndrome is the most common presence of persistent oligomenorrhea. Since
reproductive disorder with a prevalence of 4–10 % there is such a high rate of irregularity in men-
of the female population depending upon diagnos- strual periods or anovulatory periods soon after
tic criteria used. Polycystic ovarian syndrome is menarche and since there are multiple ovarian
the most common form of normal gonadotrophic cysts in normal pubertal girls, anovulatory
anovulation. PCOS has long been known to inter- symptoms and evidence of polycystic ovaries
fere with reproduction and menstrual periods and are not appropriate criteria themselves in teen-
known to cause the disheartening appearance of age girls although they are more compelling at
severe acne and hirsutism. More recently PCOS older ages. About two-thirds of girls with poly-
has been recognized as the metabolic syndrome of cystic ovarian syndrome have menstrual disor-
women based upon an insulin-resistant state, car- ders ranging from primary amenorrhea to
rying a risk of dyslipidemia leading to cardiovas- secondary amenorrhea to dysfunctional uterine
cular disease, insulin resistance and type 2 bleeding. Acne is so common in teenagers that
diabetes, and other associated manifestations of it cannot be used by itself as a criteria. However,
the insulin resistance including obstructive sleep about 60 % of girls with polycystic ovarian
apnea and hypertension. Obesity is extremely syndrome have hirsutism.
230 9 Disorders of Puberty

Measurement of anti-Müllerian hormone has presence of obesity, girls diagnosed with poly-
been suggested as helpful in the diagnosis as cystic ovarian syndrome should be evaluated
elevated levels are found in the offspring of for type 2 diabetes; a 2-h glucose tolerance test
PCOS mothers and in affected women. AMH is and/or a hemoglobin A1c determination are sug-
a reflection of the increased number of follicles gested as described in Chap. 11. Because poly-
usually found in PCOS. Normal ovarian histol- cystic ovarian syndrome should be considered
ogy is more likely related to an adrenal source the metabolic syndrome of women, screening for
of androgens. Further some but not all patients cardiovascular risk factors should be carried out;
with PCOS have elevated LH/FSH ratios. family history of cardiovascular disease, strokes
While excess androgens can cause hirsutism, or early deaths, cigarette smoking, hypertension,
acne vulgaris, and male pattern alopecia, there is a dyslipidemia, obstructive sleep apnea, and type 2
wide variety of responses in individuals to similar diabetes are all risks for cardiovascular disease in
levels of androgen. Pilosebaceous units respond polycystic ovarian syndrome.
differentially to various doses of androgen expo- Treatment of polycystic ovarian syndrome in
sure depending upon the location of the unit. adolescents depends upon the aspect to be
Sebaceous cells can metabolize dehydroepian- targeted. Obesity should always be treated by
drosterone (DHEA) in a stepwise process to lifestyle modification as the first modality. In the
androstenedione, testosterone, and dihydrotestos- treatment of acne, hirsutism, and anovulation,
terone (DHT) and thus can be considered intra- contraceptives should be invoked. The Endocrine
crine cells which convert a prohormone, DHEA, Society could find no basis to recommend one
into a potent hormone, DHT. The hair follicle has hormonal contraceptive preparation over
its base a dermal papilla while closer to the skin another. Metformin will be indicated in the
are sebaceous glands. The dermal papilla has presence of type 2 diabetes and may be used (off
enzymes that can produce dihydrotestosterone and label) in prediabetes but also may have a small
in the presence of IGF 1 hair growth progresses. but beneficial effect on decreasing weight gain
Growth hormone is required to produce sexual in some individuals. Small studies suggest that
hair demonstrating the interaction between andro- metformin works better in adolescents than
gens and the growth hormone-IGF 1 system. adults in polycystic ovarian syndrome.
Idiopathic hirsutism is not related to androgens,
and the etiology of hair growth is not yet clear for
this condition. Suggested Readings
Other conditions may have some presenting
symptoms similar to polycystic ovarian syn- 1. Main KM, Skakkebaek NE, Toppari J. Cryptorchidism
as part of the testicular dysgenesis syndrome: the envi-
drome. Nonclassic congenital adrenal hyperpla-
ronmental connection. Endocr Dev. 2009;14:167–73.
sia, most frequently 21-hydroxylase deficiency, 2. Teo AQ, Khan AR, Williams MP, Carroll D, Hughes
will lead to hirsutism and irregular menses. The IA. Is surgical exploration necessary in bilateral anor-
differential diagnosis is confused by the finding of chia? J Pediatr Urol. 2013;9(1):e78–81.
3. Rivkees SA, Hager K, Hosono S, Wise A, Li P, Rinder
elevated adrenal androgens in 20–30 % of patients
HM, Gruen JR. A highly sensitive, high-throughput
with PCOS suggesting in some patients a com- assay for the detection of Turner syndrome. J Clin
mon disorder of ovaries and adrenals. Further thy- Endocrinol Metab. 2011;96(3):699–705.
roid disease may cause irregular menstrual periods 4. Bondy CA, Turner Syndrome Study Group. Care of
girls and women with Turner syndrome: a guideline
and weight gain, and hyperprolactinemia may
of the Turner Syndrome Study Group. J Clin Endocrinol
cause irregular periods. These conditions are Metab. 2007;92(1):10–25.
discussed in other chapters and should be elimi- 5. Mauras N. Strategies for maximizing growth in
nated before the diagnosis of polycystic ovarian puberty in children with short stature. Pediatr Clin
North Am. 2011;58(5):1167–79. x.
syndrome is established.
6. Keil MF, Stratakis CA. Pituitary tumors in childhood:
Because polycystic ovarian syndrome is an update of diagnosis, treatment and molecular genetics.
insulin-resistant condition and often occurs in the Expert Rev Neurother. 2008;8(4):563–74.
Suggested Readings 231

7. Palmert MR, Dunkel L, Witchel SF. Chapter 17— Woodruff TK. Future directions in oncofertility and
Puberty and its disorders in the male. In: Sperling fertility preservation: a report from the 2011
MA, editor. Pediatric endocrinology. 4th ed. Oncofertility Consortium Conference. J Adolesc
Philadelphia, PA: Elsevier; 2014. p. 697–733. Young Adult Oncol. 2013;2(1):25–30.
8. Saenger P, Bondy CA. Chapter 16—Turner syndrome. 22. Legro RS, Arslanian SA, Ehrmann DA, Hoeger KM,
In: Sperling MA, editor. Pediatric endocrinology. 4th Murad MH, Pasquali R, Welt CK, Endocrine Society.
ed. Philadelphia, PA: Elsevier; 2014. p. 664–696. Diagnosis and treatment of polycystic ovary syndrome:
9. Rosenfield RL, Cooke DW, Radovick S. Chapter an Endocrine Society clinical practice guideline. J Clin
15—Puberty and its disorders in the female. In: Endocrinol Metab. 2013;98(12):4565–92.
Sperling MA, editor. Pediatric endocrinology. 4th ed. 23. Diamanti-Kandarakis E, Dunaif A. Insulin resistance
Philadelphia, PA: Elsevier; 2014. p. 569–663. and the polycystic ovary syndrome revisited: an
10. Terasawa E, Guerriero KA, Plant TM. Kisspeptin and update on mechanisms and implications. Endocr Rev.
puberty in mammals. Adv Exp Med Biol. 2013;784: 2012;33(6):981–1030.
253–73. 24. Fuqua JS. Treatment and outcomes of precocious
11. Palmert MR, Dunkel L. Clinical practice. Delayed puberty: an update. J Clin Endocrinol Metab. 2013;
puberty. N Engl J Med. 2012;366(5):443–53. 98(6):2198–207.
12. Harrington J, Palmert MR. Clinical review: 25. Mountjoy M, Sundgot-Borgen J, Burke L, Carter S,
Distinguishing constitutional delay of growth and Constantini N, Lebrun C, Meyer N, Sherman R,
puberty from isolated hypogonadotropic hypogonad- Steffen K, Budgett R, Ljungqvist A. The IOC consen-
ism: critical appraisal of available diagnostic tests. J sus statement: beyond the Female Athlete Triad–
Clin Endocrinol Metab. 2012;97(9):3056–67. Relative Energy Deficiency in Sport (RED-S). Br J
13. Henrichs KL, McCauley HL, Miller E, Styne DM, Sports Med. 2014;48(7):491–7.
Saito N, Breslau J. Early menarche and childhood 26. Rosenfield RL. Hirsutism and the variable response of
adversities in a nationally representative sample. Int J the pilosebaceous unit to androgen. J Investig
Pediatr Endocrinol. 2014;2014(1):14. Dermatol Symp Proc. 2005;10(3):205–8.
14. Lee Y, Styne D. Influences on the onset and tempo of 27. Rosenfield RL. Clinical review: Adolescent anovula-
puberty in human beings and implications for adoles- tion: maturational mechanisms and implications.
cent psychological development. Horm Behav. J Clin Endocrinol Metab. 2013;98(9):3572–83.
2013;64(2):250–61. 28. Voutilainen R, Jaaskelainen J. Premature adrenarche:
15. Loomba-Albrecht LA, Styne DM. Effect of puberty etiology, clinical findings, and consequences.
on body composition. Curr Opin Endocrinol Diabetes J Steroid Biochem Mol Biol. 2015;145:226–36.
Obes. 2009;16(1):10–5. 29. Styne D. Puberty. In: Gardner DG, Shoback D,
16. Carel JC, Eugster EA, Rogol A, Ghizzoni L, Palmert editors. Greenspan’s basic & clinical endocrinology.
MR, Antoniazzi F, Berenbaum S, Bourguignon JP, 9th ed. New York: The McGraw-Hill Companies, Inc.;
Chrousos GP, Coste J, Deal S, De Vries L, Foster C, 2011.
Heger S, Holland J, Jahnukainen K, Juul A, Kaplowitz 30. Styne DM, Grumbach MM. Puberty: ontogeny,
P, Lahlou N, Lee MM, Lee P, Merke DP, Neely EK, neuroendocrinology, physiology, and disorders. In:
Oostdijk W, Phillip M, Rosenfield RL, Shulman D, Williams RH, Melmed S, editors. Williams text-
Styne D, Tauber M, Wit JM. Consensus statement on book of endocrinology. Philadelphia, PA: Elsevier;
the use of gonadotropin-releasing hormone analogs in 2015.
children. Pediatrics. 2009;123(4):e752–62. 31. Finlayson CA, Styne DM, Larry Jameson J.
17. Albrecht L, Styne D. Laboratory testing of gonadal Endocrinology of sexual maturation and puberty. In:
steroids in children. Pediatr Endocrinol Rev. 2007;5 Larry Jameson J, De Groot LJ, editors. Endocrinology:
Suppl 1:599–607. adult and pediatric. 7th ed. Philadelphia, PA: Elsevier;
18. Lenz AM, Shulman D, Eugster EA, Rahhal S, Fuqua 2015.
JS, Pescovitz OH, Lewis KA. Bicalutamide and third- 32. Abreu AP et al. Central precocious puberty caused by
generation aromatase inhibitors in testotoxicosis. mutations in the imprinted gene MKRN3. N Engl J
Pediatrics. 2010;126(3):e728–33. Med. 2013;368(26):2467–75.2.
19. Shulman DI, Francis GL, Palmert MR, Eugster 33. Latronico AC, Brito VN, Carel JC. Causes, diagnosis,
EA. Use of aromatase inhibitors in children and and treatment of central precocious puberty. Lancet
adolescents with disorders of growth and adolescent Diabetes Endocrinol. 2016. pii: S2213–8587(15)
development. Pediatrics. 2008;121(4):e975–83. 00380–0.
20. Rosenthal SM. Approach to the patient: transgender 34. Gore AC et al. Executive summary to EDC-2: The
youth: endocrine considerations. J Clin Endocrinol endocrine society’s second scientific statement on
Metab. 2014;99(12):4379–89. doi:10.1210/ endocrine-disrupting chemicals. Endocr Rev. 2015;
jc20141919. 36(6):593–602.
21. Waimey KE, Duncan FE, Su HI, Smith K, Wallach H, 35. Lee JM, Wasserman R, Kaciroti N, Gebremariam A,
Jona K, Coutifaris C, Gracia CR, Shea LD, Brannigan Steffes J, Dowshen S, Harris D, Serwint J, Abney D,
RE, Chang RJ, Zelinski MB, Stouffer RL, Taylor RL, Smitherman L, Reiter E, Herman-Giddens ME.
232 9 Disorders of Puberty

Timing of puberty in overweight versus obese boys. 38. Harrington J, Palmert MR. Treatment of precocious
Pediatrics. 2016. pii: peds. 2015–0164. puberty. http://www.uptodate.com/contents/
36. Biro FM, Chan Y. Normal puberty. http://www.upto- treatment-of-precocious-puberty.
date.com/contents/normal-puberty. 39. Crowley WF, Pitteloud N. Diagnosis and treatment of
37. Harrington J, Palmert MR. Definition, etiology, and delayed puberty. http://www.uptodate.com/contents/
evaluation of precocious puberty. http://www. diagnosis-and-treatment-of-delayed-puberty.
uptodate.com/contents/definition-etiology-and- 40. Rosenfield RL. Premature adrenarche. http://www.
evaluation-of-precocious-puberty. uptodate.com/contents/premature-adrenarche.
Disorders of the Adrenal Gland
10

Disorders of the adrenal gland can lead to effects to the body than is true after birth. The fetal adre-
as disparate as abnormal sexual differentiation, nal gland has a definitive zone in the cortex which
decreased or increased growth velocity, and life- produces glucocorticoid and mineralocorticoid
threatening endocrine emergencies. Diseases can and a larger fetal zone internally. The fetal zone
arise from abnormalities within the adrenal gland lacks 3-beta hydroxysteroid dehydrogenase and
itself, from abnormalities in the control of the thus produces large quantities of dehydroepian-
adrenal gland, or from exposure to the exogenous drosterone (DHEA) and its sulfate. The steroid
steroid compounds, even if these are prescribed precursors produced by the fetal adrenal zone,
for the appropriate treatment of diseases. This DHEA and variants, are utilized by the placenta to
chapter does not discuss congenital adrenal produce estrogens, estrone, estradiol, estriol, and
hyperplasia which is found in Chap. 8. estetrol. The level of maternal estrone is an indi-
cator of the health of the fetus. In anencephaly in
which fetal ACTH is extremely decreased and the
Normal Adrenal Gland Physiology fetal adrenal zone is not stimulated, estrone levels
decrease in mother’s samples. Maternal glucocor-
The paired adrenal glands sit above the kidney ticoid production increases during pregnancy,
(which accounts for their archaic name, the supra- but the placenta through the action of 11-beta-
renal glands) and are composed of two major hydroxysteroid dehydrogenase-1 (11β-HSD1)
components derived from different embryonic tis- inactivates maternal hydrocortisone into the inac-
sues, the cortex and the medulla (Fig. 10.1). The tive form cortisone. Treatment of mothers with
adrenal cortex is derived from mesenchyme and betamethasone to enhance fetal lung maturation
produces steroid compounds synthesized from has not to date been proven to lead to long-term
the cholesterol molecule; the adrenal medulla is neurologic or other effects although long-term
derived from neural crest tissue and produces studies are ongoing to study the issue. Repeated
catecholamines. courses are recommended in some cases, and
The median eminence of the hypothalamus there is some evidence of an increase in congeni-
secretes corticotrophin-releasing factor (CRF), a tal defects, but these are not universal findings.
41-amino-acid peptide that stimulates the release The premature baby will retain fetal adrenal
of adrenocorticotropic hormone (ACTH) from function longer than the term baby, and false pos-
the anterior pituitary gland. itives in the newborn screening test for congeni-
The adrenal gland histology and function tal adrenal hyperplasia can occur because of this
change considerably between fetal life and child- immaturity. The definitive zone of the fetal adre-
hood. The fetal adrenal gland is larger in relation nal develops into the three zones of the mature

© Springer International Publishing Switzerland 2016 233


D.M. Styne, Pediatric Endocrinology, DOI 10.1007/978-3-319-18371-8_10
234 10 Disorders of the Adrenal Gland

Third ventricle Hypothalamus


Neuroendocrine
cell nuclei

CRF secreting cells

Superior
hypophyseal
artery
Stalk

Long portal
vessels
Inferior
Adrenocorticotrophs hypophyseal
artery

Secreting cells

Anterior Posterior
pituitary pituitary

Hormone Short portal


secretion vessel
Glomerulosa
ACTH
Fasciculata

Reticularis

Medulla

Aldosterone Cortisol Androgens Catecholamines

Fig. 10.1 Normal adrenal gland physiology in which to produce cortisol and androgens. Cortisol exerts gluco-
corticotropin-releasing hormone (CRH) from the hypo- corticoid effects and provides feedback inhibition on the
thalamus reaches the pituitary gland through the hypo- pituitary gland and hypothalamus to suppress CRH and
physiotropic portal system to cause ACTH secretion. ACTH secretion until a normal equilibrium is reached
ACTH interacts with its receptors on the adrenal glands

adrenal gland cortex: the zona glomerulosa, zona The periventricular nucleus of the hypothala-
fasciculata, and the zona reticularis going from mus produces and secretes corticotrophin-
the outer most cortex toward the medulla. The releasing factor (CRF) which stimulates the
zona glomerulosa predominantly produces min- pituitary production and secretion of adrenocor-
eralocorticoids, the zona fasciculata glucocorti- ticotropin or ACTH. Both CRF and vasopressin
coids, and the zona reticularis adrenal androgens are secreted at times of stress and both can
although there is overlap between the zones. stimulate ACTH secretion. The stress response
Normal Adrenal Gland Physiology 235

characteristically relates to physical or meta- are saturable transporters that can bring them
bolic stress. Psychological stress can also trig- across the blood brain barrier.
ger ACTH release. Other factors enhancing ACTH is a polypeptide hormone which acts
ACTH secretion are hemorrhagic shock and through a G protein seven-transmembrane domain
hypovolemia. The hypothalamic–pituitary axis receptor, the MC2R receptor. ACTH stimulates
response to stress is essential in insuring appro- the uptake of LDL cholesterol into the adrenal
priate cardiovascular response to angiotensin II gland, to a smaller extent the production of choles-
and catecholamines and on the other hand inhib- terol, and ultimately the production of glucocorti-
iting excessive activation of the immune sys- coids in the zona fasciculate through several steps
tem. Inflammatory cytokines including IL-1β, (Fig. 10.2). ACTH causes cholesterol esters to be
IL-6, and TNF mediate the adrenal response to converted to free cholesterol by cholesterol ester
stress in part due to intraadrenal direct stimula- hydrolase and enables their transport into the mito-
tion of glucocorticoid production and in part chondria by its effect on the steroidogenic acute
due to stimulation of CRF and ACTH secretion. regulatory protein (StAR). Within the mitochon-
Glucocorticoids are effective in suppressing the dria the side-chain cleavage enzyme (P450 SCC)
proinflammatory cytokine response characteris- converts cholesterol to pregnenolone which is con-
tic of autoimmune, inflammatory, and allergic verted to progesterone by 3-beta hydroxysteroid
diseases by directly inhibiting the activity of dehydrogenase. Progesterone is 17-hydroxylated
activated T cells, monocytes, and macrophages by 17,20-lyase (P450 C17) and the resulting
and by decreasing the production of proinflam- 17-hydroxyprogesterone is the precursor to
matory cytokines. The lytic effects of glucocor- 21-hydroxylase (P4 50 C 21). 17-OHP is extremely
ticoids are used in the treatment of leukemia and elevated in 21-hydroxylase deficiency congenital
other tumors. However, in sepsis and various adrenal hyperplasia leading to the diagnosis of the
other forms of stress, there can be a disassocia- condition. 17-Hydroxyprogesterone is 21-hydrox-
tion from appropriate adrenal stimulation lead- ylated to produce 11-deoxycortisol. Finally
ing to critical illness-related corticosteroid 11-deoxycortisol, also known as compound S, is
insufficiency (CIRCI) of the HPA axis. 11-beta hydroxylated (P4 50 C11 beta) to produce
Corticotropin or ACTH is cleaved from the pre- cortisol (also called compound F or hydrocorti-
cursor molecule, pro-opiomelanocortin (POMC), sone). Cortisol through negative feedback inhibi-
which also plays a role in appetite regulation tion of the central nervous system (CNS) reduces
(see Chap. 13), after posttranslational modifica- the secretion of CRF and therefore ACTH until
tions. ACTH is released in a pulsatile manner with equilibrium is reached.
a diurnal variation of increased values prior to Cortisol is a glucocorticoid that stimulates
awakening in the morning which is controlled by gluconeogenesis, maintains blood pressure, and
a biological “clock” in the suprachiasmatic affects mood. The secretory rate of cortisol var-
nucleus (SCN) of the ventral hypothalamus. ies depending upon the method of measure-
Other products of the cleavage of POMC that ment. Stable isotope-dilution technique
exert effects on the central nervous system and employing high-performance liquid chromatog-
elsewhere in the body include α-melanotropin raphy–mass spectrometry revealed a secretory
(α-melanocyte-stimulating hormone, or α-MSH) rate of 6.8 ± 1.9 mg/m2/day, while the use of
which causes pigmentation characteristic of states stable isotope dilution using gas chromatogra-
where there is increased ACTH secretion, phy–negative ion chemical ionization mass
β-melanotropin (β-MSH), N-terminal peptide of spectrometry revealed a secretory rate of
pro-opiomelanocortin (NPP, or pro-γ-MSH), 4.8 ± 0.6 mg/m2/24 h in male children and
γ-melanotropin (γ-MSH), corticotropin-like inter- 4.4 ± 0.5 mg/m2/24 h in female children. Presently
mediate peptide (CLIP), β-lipotropin (β-LPH), most clinicians utilize a normal secretory rate of
lipotropin gamma (γ-LPH), β-endorphin, and 6–8 mg/m2/day as a starting point for replacement
[Met]enkephalin. TNF-α and IL-1β are inflamma- therapy and stress dosing with adjustments made
tory cytokines that increase ACTH secretion; there upon the patient’s response: oral glucocorticoid
236 10 Disorders of the Adrenal Gland

Cholesterol
P450SCC
Δ5
Pregnenolone

3β-HSD
P450c17
Progesterone (17α-OH)

P450c17
(17α-OH) Δ5 17-OH
Pregnenolone
P450c17
17-OH (17,20 lyase)
Progesterone
P450c21
P450c21 (21-OH)
(21-OH) 17βHSD-111
Dehydroepiandrosterone Androstenediol
(DHEA)
Deoxycortico-
Sterone 3β-HSD
(DOC) 17βHSD-111
Deoxycortisol (S) Δ4
P450c11β P450c11 Androstenedione Testosterone
(11β-OH) (11β-OH)
P450aro P450aro 5α-
P450c11 AS reductase
(18-OH) Cortisol (tissue)
Estrone Estradiol
17βHSD-1

Corticosterone (B)
Dihydrotestosterone
P450c11 AS
(18-OH)

18-OH Corticosterone
P450c11 AS
(18-oxidase)

Aldosterone

Mineralocorticoid Glucocorticoid Sex steroids

Zona glomerulosa Zona fasciculata Zona reticularis

Fig. 10.2 Normal adrenal gland physiology as gland in which much of the activity that occurs is noted
described in the text. Enzymes are noted in blue, major in bold at the bottom
products are noted in red, and the zone of the adrenal

dosage is about double these values. Most but Steroid and thyroid receptors are members of
not all studies show that increased BMI and a “superfamily” including receptors for steroid
waist–hip ratio and increased visceral fat hormones (hydrophobic molecules) as well as
increase the ACTH response to stimulatory test- thyroid hormones, fatty acids, prostaglandins,
ing and increase AM cortisol and urinary free and leukotrienes (see Fig. 1.5). Unbound
cortisol. glucocorticoid receptors (GR) reside in the
Normal Adrenal Gland Physiology 237

cytosol in complexes with various proteins and pituitary from these conditions leads to
including heat-shock protein 90 (hsp90), heat increased ACTH which causes the adrenal gland
shock protein 70 (hsp70), and FKBP52 (FK506- to produce excessive amounts of products
binding protein 52). Cortisol passively enters the through the remaining operative pathways which
cells where it binds to the glucocorticoid receptor in the most common forms of CAH is androgens
(GR) also known as NR3C1 (nuclear receptor which virilize a female fetus. Congenital adrenal
subfamily 3, group C, member 1) releasing the hyperplasia is named for the large size of the
hsps. The cortisol–receptor complex homodi- adrenal glands which occurs because of the
merizes and translocates through active trans- excessive stimulation of ACTH. Even though the
port to the nucleus, where it binds to glands are hyperplastic in congenital adrenal
DNA-responsive elements to activate gene tran- hyperplasia, they do not produce cortisol, unlike
scription in a process known as transactivation. the bilateral adrenal hyperplasia of Cushing dis-
In other cells the cortisol–receptor complex can ease. It is important not to confuse the adrenal
further complex with other transcription factors to hyperplasia of Cushing disease with congenital
suppress the expression of genes that ordinarily adrenal hyperplasia.
are unregulated by these transcription factors in a Cortisol circulates mostly (80–90 %) protein
process known as transrepression. bound to cortisol-binding globulin (CBG or trans-
Cortisol has many metabolic actions in the cortin), which serves as a reservoir to smooth out
body. Cortisol enhances gluconeogenesis and the peaks and valleys of cortisol secretion. While
inhibits production of interleukin (IL) 12, inter- cortisol binds to CBG, synthetic glucocorticoids
feron (IFN)-gamma, IFN-alpha, and tumor- such as prednisolone or dexamethasone bind
necrosis factor (TNF)-alpha by antigen-presenting poorly to CBG partially explaining their increased
cells (APCs) and T helper (Th)1 cells, while potency. If excess glucocorticoid is produced, as
upregulating IL-4, IL-10, and IL-13 by Th2 cells. in Cushing disease, free cortisol will overwhelm
Cortisol enhances glycogenolysis by activating the available CBG causing increasing free cortisol
glycogen phosphorylase, so that epinephrine can levels bringing about excessive glucocorticoid
exert its effect on glycogenolysis. Chronic expo- effects and spillover into the urine as urinary free
sure to elevated cortisol as in Cushing syndrome cortisol. Like TeBG, CBG increases with
leads to proteolysis and muscle wasting and increasing estrogen and decreases with increas-
weakness and a decrease in bone formation. ing androgen. Autosomal dominant or autoso-
Excess cortisol will inhibit wound healing due to mal recessive CBG deficiency (#611489
effects on connective tissue. CORTICOSTEROID-BINDING GLOBULIN
Cortisol is metabolized in the liver by reduc- DEFICIENCY) may be asymptomatic as free
tion to cortisone, oxidization to 17-ketosteroids, cortisol remains normal, but complaints of fatigue
or hydroxylation to 6-beta-hydroxycortisol. The or hypotension are reported.
metabolites are excreted in the urine as they The weak adrenal androgen, dehydroepian-
become water soluble by conjugation with sul- drosterone (DHEA), is produced in the zona retic-
fate or glucuronic acid. Metabolism is increased ularis. Production is dependent on the presence of
with elevated thyroid hormone. The kidney also ACTH, as without ACTH no significant amount
metabolizes cortisol with 11-beta-hydroxysteroid of adrenal androgens will be produced. But DHEA
dehydrogenase type 2 which converts cortisol to is produced in increased quantities at the time of
cortisone. Urinary 17-hydroxycorticosteroids normal puberty; this is not related to a change in
(17-OHCS) measure both cortisol and its urinary ACTH secretion. The control of this increase in
metabolites. adrenal secretion, known as adrenarche, is poorly
Each of these enzymatic processes can fail characterized but does not occur in the absence of
due to loss-of-function mutations leading to ACTH. It is possible that some undiscovered pitu-
various types of congenital adrenal hyperplasia. itary hormone or intra-adrenal processes mediate
The lack of cortisol feedback to the hypothalamus adrenarche. DHEA is produced from ∆5 17-OH
238 10 Disorders of the Adrenal Gland

pregnenolone by the 17,20-lyase activity of P450 group C, member 2 (NR3C2) in a manner similar
C17. DHEA serves as a precursor for the more to cortisol mostly in the kidney collecting ducts
potent androgens androstenedione, androstene- but has effects on the colon and skin to a minor
diol, and ultimately testosterone from peripheral degree. Cortisol can interact with the aldosterone
conversion (see Chap. 9). These androgens can be receptor and excessive cortisol exerts a mineralo-
aromatized to estrogen in the periphery. DHEA is corticoid effect. This effect is decreased by local
weakly bound to sex hormone binding while conversion of cortisol to the inactive molecule cor-
DHEAS is not bound to that protein. tisone by 11-beta-hydroxysteroid dehydrogenase 2
Aldosterone, the main mineralocorticoid of the decreasing the effect of cortisol on aldosterone-
adrenal cortex, under normal physiologic condi- sensitive tissue.
tions is regulated by the renin–angiotensin axis Aldosterone interacts with its receptor to stim-
and less by ACTH. If there is ACTH deficiency as ulate the active reabsorption of sodium and chlo-
in secondary or tertiary adrenal insufficiency, ride from the proximal convoluted tubule and
mineralocorticoid function will still remain nor- excretion of K by the sodium–potassium-
mal. However, mineralocorticoids can be secreted activated adenosine triphosphatase pump (Na(+)–
in response to abnormally high concentrations of K(+)-ATPase; Na(+)–K+ pump), as, to a lesser
ACTH, as found in ectopic ACTH-secreting extent, does its precursor, deoxycorticosterone
tumors. Likewise vasopressin not only stimulates (DOC), another potent mineralocorticoid. When
ACTH secretion but may also exert a brief stimu- DOC is produced in excess, as in 17-OH defi-
lation of aldosterone secretion. With lowered total ciency congenital adrenal hyperplasia (CAH), an
body sodium or hypotension or just simply stand- increased mineralocorticoid effect will be noted,
ing up, renin, an enzyme produced in the juxta- but renin will be suppressed, as will aldosterone.
glomerular apparatus of the kidney, acts on The serum concentrations of sodium and potas-
angiotensinogen to form angiotensin I; angioten- sium reveal much about mineralocorticoid func-
sin I is transformed by the converting enzyme of tion: without mineralocorticoid effect, serum
blood and lung to form the octapeptide, angioten- sodium decreases and serum potassium increases,
sin II. Angiotensin II (and its metabolite angioten- whereas in states of excess mineralocorticoid, total
sin III) stimulates aldosterone production and body sodium increases, serum sodium increases to
secretion from the zona glomerulosa of the adre- high normal, and blood pressure increases while
nal cortex. Elevated potassium will also stimulate potassium decreases. DOC also is produced in the
aldosterone production. zona fasciculata, an ACTH-responsive region of
Aldosterone circulates with about 67 % bound the adrenal gland that produces mainly cortisol.
(17 % bound to transcortin and 47 % to albumin) Thus, when ACTH is extremely elevated, as found
with remaining 36 % circulating as free hormone. in ectopic ACTH syndrome, as indicated above,
Like cortisol, aldosterone is metabolized in the mineralocorticoid increases and causes hypokale-
liver by reduction, oxidization, and hydroxyl- mia, possibly with alkalosis, and increased blood
ation with the major urinary metabolite being pressure because of sodium retention. The new-
tetrahydroaldosterone glucuronide. Metabolism born is relatively insensitive to aldosterone.
is decreased in liver disease. Hyponatremia and hyperkalemia due to salt-
Similar to cortisol, aldosterone passively losing congenital adrenal hyperplasia do not
enters the cells where it binds to the aldosterone develop until 3–5 days after birth which may relate
receptor or nuclear receptor subfamily 3, group C, to the lack of necessity for aldosterone action to
member 2 (NR3C2). The aldosterone–receptor maintain normal sodium and potassium during
complex homodimerizes and translocates through these first several days.
active transport to the nucleus, where it binds to Serum ACTH normally increases in the morn-
DNA-responsive elements to activate gene tran- ing before awakening, at about 4:00–6:00 a.m., in
scription in a process known as transactivation. a person on a customary daily schedule, and serum
Aldosterone interacts with its specific aldoste- cortisol increases in a couple of hours thereafter in
rone receptor or nuclear receptor subfamily 3, response. Serum concentrations of both ACTH
Measurement of Steroids 239

and cortisol decrease in the afternoon and evening It is essential that the values determined by any
as part of the normal circadian rhythm of adrenal method be referenced to standards for the age of
activity. The circadian rhythm of ACTH and corti- the child. Since not all laboratories have pediatric
sol is not present in the newborn. It generally takes standards, it is important to choose a laboratory
up to 7–8 weeks for the normal circadian rhythm that does have standards for the correct age of
to develop. If an individual changes to a different your patient. Thus it is generally necessary to
sleep–wake cycle than they are used to, there will send samples for analysis to larger national labo-
be a disorder in the circadian rhythm; after a period ratories that have the correct techniques and the
of approximately 21 days, the circadian rhythm correct pediatric standards.
will adjust to the new schedule; thus, a person A urinary free cortisol determination is quite
waking at night and sleeping during the day will specific for cortisol. Alternative methods measure
have the peak of ACTH and cortisol phase shifted 17-hydroxycorticosteroid (17-OHCS) which is a
to increase just before awakening at night. The metabolite of cortisol. Normal urinary free corti-
delay in this adjustment however can lead to physi- sol is 11 ± 5 mg/m2/day and clearly relates to
ologic stress in the body; it is likely one of the rea- body size. An older test, 17-ketogenic steroids, is
sons for “jet lag.” rarely invoked nowadays as an indication of uri-
Because ACTH concentrations vary consider- nary glucocorticoid metabolites. Urinary metab-
ably due to the episodic nature of ACTH secretion, olites of DHEA and DHEAS are measured as
a single-serum ACTH determination may not be a 17-ketosteroids (17 KS). Thus 17-ketosteroids
valid reflection of the ACTH–cortisol axis. Serum measure urinary adrenal androgens but do not
cortisol concentrations also rise episodically but measure the more potent testosterone.
are more stable than those of ACTH because of Measurement of ACTH presents technical
protein binding of cortisol by CBG and other pro- problems and should be carried out in a reliable
teins. Cortisol can be quickly elevated by stress; laboratory. Plasma needs to be handled in a sensi-
unless serum samples for cortisol are obtained tive manner and kept cold.
under relaxed conditions or unobtrusively obtained ACTH-stimulation tests are used to determine
through an indwelling intravenous catheter, a high adrenal secretory ability and to diagnose congen-
value may be specious. Serum cortisol concentra- ital adrenal hyperplasia. The standard tests utilize
tions, when abnormally elevated for the time of a sample 30 and 60 min after the intravenous
day, can be considered a reflection of stress or infusion of synthetic ACTH (1–24). The standard
excessive secretion (as might be seen in Cushing dose is 15 μg/kg in children up to 2 years of age
disease) and the difference between the two is of and 0.25 mg for children older than 2 years, but
great importance. Thus, in Cushing disease evening these are maximal doses and it is suggested that
cortisol levels may match morning cortisol levels these high doses do not reveal whether the adre-
leading to an overall increase in cortisol effect. nal glands can actually mount increased secre-
tory response during stress. A lower dose is
suggested to be better able to determine whether
Measurement of Steroids the adrenal gland is likely to function adequately
in emergency situations. Thus a 1 μg dose is sug-
There are various methods available to measure gested to evaluate the adrenal gland response to
steroids in urine and plasma. Radioimmunoassays determine if cortisol secretion will rise in stressful
are subject to cross-reaction between various ste- situations or whether exogenous glucocorticoids
roid molecules which can change results. Thus should be administered at times of stress. While
the most accurate method of determining steroid there is controversy over the ideal peak of corti-
levels in the plasma is the use of high-performance sol after ACTH stimulation to eliminate risk of
liquid chromatography (HPLC)/tandem mass adrenal insufficiency, a peak value of 18–20 μg/dl is
spectroscopy (MS/MS) which is a more expen- usually considered adequate. There remains con-
sive method usually available only in national troversy over whether the high or low dose ACTH
laboratories but is a much more specific method. test is the superior method.
240 10 Disorders of the Adrenal Gland

Other methods may be invoked to determine helpful for the parents of a child with congenital
pituitary or adrenal secretory ability. Insulin- adrenal hyperplasia whenever they need to visit
induced hypoglycemia is a dangerous test but an emergency department to tell emergency
when carefully performed can indicate the ability providers that the child has Addison’s disease to
of the pituitary gland to secrete adequate ACTH ensure appropriate treatment. Tuberculous infec-
and the adrenal gland to respond by increasing tion of the adrenal gland was more common in
cortisol secretion. Insulin-induced hypoglycemia previous generations and gave rise to most of the
is considered the “gold standard” for determining earlier reports of Addison’s disease. Autoimmune
growth hormone secretory ability as well. disease is responsible for the majority of cases of
Metyrapone blocks 11-hydroxylase and tempo- Addison’s disease at present, and often other
rarily renders the adrenal gland incapable of pro- associated autoimmune disorders are found as
ducing cortisol leading to an increase in ACTH autoimmune polyendocrinopathy syndromes
due to the decrease in negative feedback inhibi- (see below). Antibodies to adrenal enzymes
tion by cortisol. A rise in 11-deoxycortisol, the (antibodies to P450c2, the most commonly posi-
precursor to the block induced by metyrapone, tive, or P450scc and P450c17 are tested) are
indicates pituitary ACTH secretory ability; this measurable. Because Addison’s disease causes
test cannot be utilized in primary adrenal failure progressive destruction of the adrenal gland,
since the adrenal gland will not be able to secrete some function may be retained and a patient may
11-deoxycortisol. present with glucocorticoid deficiency and only
later develop mineralocorticoid deficiency or
more rarely mineralocorticoid precedes gluco-
Disorders of the Adrenal Cortex corticoid deficiency.

Hypoadrenal States Autoimmune Polyglandular


Endocrinopathy
The most common causes of decreased cortisol Type 1 autoimmune polyglandular syndrome
production are the congenital adrenal hyperplasia (*240300 AUTOIMMUNE POLYEN-
(CAH) syndromes discussed in Chap. 8. In these DOCRINOPATHY SYNDROME, TYPE I)
conditions, ACTH is elevated due to reduced pro- includes Addison’s disease as well as hypopara-
duction of cortisol and the ensuing lack of nega- thyroidism, pernicious anemia, hypoparathy-
tive feedback inhibition on ACTH secretion. roidism, hypogonadism, and type IA diabetes
Increased cortisol secretion does not occur in mellitus and may include autoimmune pituitary
response to the elevated ACTH because of enzy- defects. This condition is also known as autoim-
matic defects in the adrenal gland which interfere mune polyendocrinopathy-candidiasis-
with the production of cortisol and abnormalities ectodermal dysplasia (APECED). Chronic muco-
of adrenal sex steroids and/or mineralocorticoids cutaneous candidiasis may be a helpful physical
occur. finding as it is often the first condition noted.
Other findings may be found in the gastrointesti-
Addison’s Disease (Fig. 10.3) nal system (diarrhea or chronic atrophic gastritis)
Addison’s disease is a complete primary adrenal and integument (vitiligo and alopecia). Antibodies
gland failure, and an affected patient exhibits the related to the affected organ system may be
results of glucocorticoid and mineralocorticoid detected in the blood. APECED is caused by
deficiency. Addison’s disease is rare in pediat- mutations in the “autoimmune regulator” (AIR)
rics, but the same findings may be found in some gene. Type I autoimmune polyglandular syn-
salt-losing forms of CAH or other congenital drome is characteristically inherited in an autoso-
defects of the adrenal gland. Because Addison’s mal recessive manner, but there are reports of
disease is well known and the details of congeni- autosomal dominant inheritance. Adrenal insuffi-
tal adrenal hyperplasia are less known, it can be ciency itself can cause elevated TSH suggesting
Disorders of the Adrenal Cortex 241

Third ventricle Hypothalamus


Neuroendocrine
cell nuclei

CRF secreting cells

Superior
hypophyseal
artery
Stalk

Long portal
vessels
Inferior
Adrenocorticotrophs hypophyseal
artery

Secreting cells

Anterior Posterior
pituitary pituitary

Hormone Short portal


secretion vessel
Glomerulosa
ACTH
Fasciculata

Reticularis

Medulla

Aldosterone Cortisol Androgens Catecholamines

Fig. 10.3 The hypothalamic–pituitary–adrenal axis in secretion. ACTH rises in the presence of low values of
primary adrenal failure. In the absence of cortisol produc- cortisol establishing the diagnosis of primary adrenal
tion, there is no feedback inhibition on CRF or ACTH insufficiency

hypothyroidism, but this finding resolves with glu- addition to adrenal insufficiency. Other findings
cocorticoid therapy; this may make the evaluation may include alopecia, ophthalmologic findings
of autoimmune disease more complex. (cataracts, keratoconjunctivitis, band keratopathy),
Type 2 autoimmune polyglandular syndrome chronic candidiasis of mucosa, skin, and nails,
(269200 SCHMIDT SYNDROME), or Schmidt candidal granuloma, thymoma, thymic dysplasia,
syndrome, is usually found in middle-aged T-lymphocyte deficiency, hypothyroidism,
women and includes thyroiditis and/or diabetes hyperthyroidism, type IA diabetes mellitus, type
mellitus type 1 A and, later, ovarian failure, in 2 diabetes, pernicious anemia, myxedema, tetany
242 10 Disorders of the Adrenal Gland

and seizures, laryngitis, chronic pulmonary mately leading to dementia) with glucocorticoid
disease, pancreatic insufficiency, steatorrhea, deficiency; some individuals also demonstrate
chronic hepatitis, cirrhosis, splenic agenesis, and mineralocorticoid deficiency. Blindness, hyper-
iron-deficiency anemia. Hypoparathyroidism is pigmentation, degenerative neurologic disorder,
not part of the syndrome. While candidiasis is not slurred speech, spastic paraplegia, and peripheral
classically described as part of type II autoim- neuropathy are seen; ultimately impotence and
mune polyglandular syndrome, OMIM lists can- sphincter disturbances develop as well as limb
didiasis as an association in this condition. and truncal ataxia. Primary adrenal failure and
Inheritance varies and may be autosomal reces- gonadal failure develop in childhood in the usual
sive, autosomal dominant, or multifactorial. A presentation of the more severe childhood or
relation is seen to human leukocyte antigen (HLA) cerebral form while the milder form develops in
type DR32 and DR4 and type II autoimmune adults, termed adrenomyeloneuropathy. Elevated
polyglandular syndrome. long-chain fatty acids are found in plasma, fibro-
Type III autoimmune polyglandular syndrome blasts, amniocytes, and chorionic villi because of
involves autoimmune hypothyroidism and possibly peroxisomal lignoceroyl-coenzyme A (CoA)
type 1 diabetes but not usually adrenal disease. ligase deficiency due to mutations in the ATP-
binding cassette, subfamily D, member 1 gene
Wolman Disease (ABCD1). On radiographic examination, atrophy
A rare form of mutation of the gene for choles- of pons and cerebellum are seen on CT scan. A
terol esterase that limits cholesterol entry into the defect in the metabolism of very-long-chain fatty
steroidogenic pathways is described. Wolman acids is noted, and serum hexacosanoic acid
disease (*278000 WOLMAN DISEASE 10q24- (c26) is elevated, leading to an abnormally high
q25 or primary xanthomatosis) is an autosomal ratio of c26/c22 fatty acids. Newborn screening
dominant disorder of adrenal insufficiency that programs increasingly test for this condition.
manifests with vomiting, diarrhea, steatorrhea, This condition must always be in the differential
hepatosplenomegaly, hepatic fibrosis, esophageal diagnosis of non-autoimmune adrenal failure.
varices, and intestinal malabsorption. The condi-
tion leads to cholesterol and triglyceride accumu- Congenital Adrenal Hypoplasia or AHC
lation in all cells of the body and is ultimately AHC (*300200 ADRENAL HYPOPLASIA,
fatal although bone marrow transplantation has CONGENITAL; AHC at Xp21.3-p21.2) may
shown promise in treatment of some. Diffuse par- cause an Addison-like condition, occurring in an
ticulate adrenal calcifications are seen on radio- X-linked pattern, with histologic findings of cyto-
graph. Laboratory evaluation reveals acid megaly; this may be accompanied by hypogonad-
cholesteryl ester hydrolase deficiency, foam cell otropic hypogonadism. The X-linked form is due
infiltration in the bone marrow as well as dissem- to mutations in the NR0B1 gene encoding DAX-1
inated in other organs, vacuolated blood lympho- gene and may occur with glycerol kinase defi-
cytes, and normal or moderately elevated plasma ciency, ornithine transcarbamylase (OTC) defi-
lipids with hypercholesterolemia. There are xan- ciency, and muscular dystrophy. An autosomal
thomatous changes in the liver, adrenal, spleen, pattern of inheritance is described as well (202150
lymph nodes, bone marrow, small intestine, ADRENAL HYPOPLASIA, CONGENITAL,
lungs, and thymus and slight changes in skin, WITH ABSENT PITUITARY LUTEINIZING
retina, and CNS. HORMONE), which is associated with the
appearance of a miniature adult cortex and com-
Adrenoleukodystrophy plete lack of the fetal cortex at birth; this also is
Adrenoleukodystrophy, called bronze Schilder associated with the absence of pituitary LH.
disease in the past (*300100 ADRENOLEU- Patients have findings of adrenal insufficiency
KODYSTROPHY; ALD at Xq28), is an X-linked including glucocorticoid deficiency and miner-
disorder that combines degeneration of the white alocorticoid deficiency. Treatment is accom-
matter of the brain (causing CNS changes, ulti- plished with replacement of both hormones.
Disorders of the Adrenal Cortex 243

Congenital Unresponsiveness to ACTH corticotropin-releasing factor; when any form of


Congenital unresponsiveness to ACTH is due to hypothyroidism is diagnosed, there must be
homozygous or compound heterozygous muta- consideration of associated glucocorticoid
tion in the gene encoding melanocortin-2 receptor deficiency.
(MC2R; 607397), which is also the adrenocorti- Congenital defects of pituitary development
cotropin receptor (ACTHR) and presents with lead to multiple pituitary hormone deficiencies
glucocorticoid deficiency but not mineralocorti- which can include ACTH deficiency as noted in
coid deficiency. This receptor is a G protein-cou- Chap. 3. Isolated ACTH deficiency is exceed-
pled cell membrane receptor which exerts effects ingly rare but reported in an autosomal recessive
through the activation of adenyl cyclase which pattern (#201400 ICD+ ACTH DEFICIENCY,
leads to increased synthesis of cAMP (see ISOLATED; IAD). Another rare condition mani-
Chap. 1, Fig. 1.4). This may be found in an auto- festing glucocorticoid deficiency is pro-
somal recessive pattern in familial glucocorticoid opiomelanocortin deficiency (#609734
deficiency type I (#202200 GLUCOCORTICOID PRO-OPIOMELANOCORTIN DEFICIENCY)
DEFICIENCY 1; GCCD). Familial glucocorti- which is characterized by pale skin color and red
coid deficiency type 2 is due to a mutation in the hair associated with insatiable appetite and
melanocortin-2 receptor accessory protein or remarkable early-onset weight gain. POMC is the
MRAP also in an autosomal recessive pattern precursor of ACTH and is also an important factor
(#607398 GLUCOCORTICOID DEFICIENCY in the control of appetite as described in Chap. 13.
2; GCCD2). Symptoms are those of glucocorti- Proprotein convertase 1/3 deficiency (*162150
coid insufficiency without abnormal electrolytes PROPROTEIN CONVERTASE, SUBTILISIN/
or dehydration; due to elevated ACTH levels, KEXIN-TYPE, 1; PCSK1), likewise another
hyperpigmentation is prominent. Familial gluco- rare disorder, leads to early-onset and severe
corticoid deficiency may combine with alacrima, obesity, but because the processing of POMC is
achalasia, and autonomic symptoms to cause abnormal, adrenal insufficiency or ACTH defi-
Allgrove syndrome (#231550 ACHALASIA- ciency can occur.
ADDISONIANISM-ALACRIMA
SYNDROME; AAA at 12q13) due to a mutation Miscellaneous
in the ALADIN gene (*605378 ALADIN; Adrenal hemorrhage in the newborn may lead to
AAAS) in an autosomal recessive pattern. Some calcifications within the adrenal gland and a flank
individuals with Allgrove syndrome also demon- mass and if severe and widespread leads to com-
strate mineralocorticoid deficiency. promise of adrenal function. Cysts and tumors of
the adrenal cortex and medulla also may lead to
Secondary or Tertiary Adrenal calcifications within the adrenal gland. The
Insufficiency (Fig. 10.4) Waterhouse–Friderichsen syndrome of adrenal
ACTH deficiency from the pituitary gland or hemorrhage associated with meningococcemia
corticotropin-releasing factor deficiency from the or other types of bacterial sepsis is rare but devas-
hypothalamus will lead to glucocorticoid defi- tating, leading to shock and, if untreated, death.
ciency and androgen deficiency of the adrenal Meningococcemia is often associated with men-
gland but not mineralocorticoid deficiency. Any ingitis. Smith–Lemli–Opitz syndrome (270400
of the tumors noted in Chaps. 3, 5, and 9 can ICD+ SMITH-LEMLI-OPITZ SYNDROME;
cause these problems although growth hormone SLOS) is a dysmorphic condition which is some-
and other pituitary hormones are usually affected times associated with adrenal insufficiency due
more frequently than ACTH. Because of coexist- to mutations in the delta-7-dehydrocholesterol
ing thyrotropin-releasing factor or TSH defi- reductase gene; ambiguous genitalia and cryptor-
ciency due to such tumors, treatment of the chidism may be found in the male as well.
hypothyroidism may bring about adrenal crisis HIV infection can cause adrenal dysfunction
due to unrecognized deficiency of ACTH or up to adrenal insufficiency by direct glandular
244 10 Disorders of the Adrenal Gland

Third ventricle Hypothalamus


Neuroendocrine
cell nuclei

CRF secreting cells

Superior
hypophyseal
artery
Stalk

Long portal
vessels
Inferior
Adrenocorticotrophs hypophyseal
artery

Secreting cells

Anterior Posterior
pituitary pituitary

Hormone Short portal


secretion vessel
Glomerulosa
ACTH
Fasciculata

Reticularis

Medulla

Aldosterone Cortisol Androgens Catecholamines

Fig. 10.4 The hypothalamic–pituitary–adrenal axis in will happen if there is an absence of CRF in tertiary
secondary (shown here) or tertiary adrenal failure. In the hypercortisolism when low ACTH and cortisol values will
absence of ACTH, there is no stimulation of the adrenal result (tertiary hypercortisolism is not demonstrated on
gland to produce cortisol and thus ACTH and cortisol val- the slide but would be represented by an X over the hypo-
ues are both low in secondary hypocortisolism. The same thalamus rather than over the pituitary gland)

invasion by infectious organisms, infiltrate or Symptoms of Adrenal Insufficiency


inflammation associated with chronic viral infec- Acute adrenal crisis may be the first indication
tion, or drug effects. that the child has adrenal insufficiency and may
Chemotherapy often includes the use of be fatal if not appropriately treated with correct
prednisone or dexamethasone, both of which can fluids, corticosteroids, and glucose. In primary
cause ACTH suppression leading to secondary adrenal gland failure there is characteristically
adrenal insufficiency. loss of glucocorticoid and mineralocorticoid
Disorders of the Adrenal Cortex 245

function. However, acute adrenal crisis may Glucose/insulin infusions or oral potassium-
alternatively occur with abrupt discontinuation of retaining resin may be used to bring about a rapid
chronic glucocorticoid therapy (10–14 days can decrease in extremely elevated serum K if ECG
establish adrenal suppression) without involve- changes (peaked T waves) suggest such severe
ment of mineralocorticoid deficiency. Symptoms hyperkalemia.
of acute adrenal insufficiency include hypoglyce- Deficiency of pituitary ACTH (secondary
mic seizures, abdominal pain, fever, vomiting, adrenal insufficiency) or hypothalamic CRF (ter-
acidemia, hypotensive shock and death, and if it tiary adrenal insufficiency) may occur from a
occurs with mineralocorticoid failure there will be congenital defect or a hypothalamic–pituitary
hyponatremia, hyperkalemia, and hypochloremia. tumor; usually an associated deficiency of other
Symptoms of chronic glucocorticoid deficiency anterior pituitary hormones is noted (see Chap.
include weight loss or lack of adequate weight 3). Vasopressin deficiency may accompany the
gain, weakness and lethargy, fatigue, hypoglyce- ACTH deficiency and diabetes insipidus may be
mia, and anorexia, whereas mineralocorticoid masked due to the inability to excrete a water
deficiency is heralded by hyponatremia with load due to glucocorticoid deficiency as described
hyperkalemia, hypotension, low-voltage electro- above. Thus with glucocorticoid treatment,
cardiogram (ECG) with small heart on radio- diabetes insipidus might develop clinically.
graph, tachycardia, and acidosis. Mineralocorticoid function should be normal in
Glucocorticoid deficiency may lead to an secondary or tertiary adrenal insufficiency.
inability to excrete a water load indicated by a
decrease in urine output, thereby lowering serum Diagnosis of Hypoadrenal States
sodium and osmolality by dilutional effects even Laboratory evaluation of primary adrenal failure
without mineralocorticoid deficiency. will demonstrate elevated ACTH concentrations
ACTH rises in primary adrenal failure and at any time of the day. Serum cortisol measures
metabolism of ACTH to melanocortin leads to CBG-bound cortisol and free cortisol. Decreased
hyperpigmentation of exposed skin, especially of cortisol concentrations when measured in the
flexural or injured surfaces, as well as hyperpig- morning hours (because values are already low in
mentation of the gum line. Monilial lesions of the the evening, measurement then may not
skin or nails will occur in the syndromes associ- differentiate between normal and abnormal val-
ated with mucocutaneous candidiasis and adrenal ues) (Fig. 10.2) suggest deficient glucocorticoid
insufficiency, which combines an immune defect secretion. Reduced cortisol reserve is noted on
with an autoimmune disorder of the adrenal ACTH-stimulation testing or insulin-induced
glands. Salt craving may be noted on questioning hypoglycemia (we do not recommend insulin-
if mineralocorticoids are affected as the child may induced hypoglycemia testing if the physician has
add a remarkable amount of salt to his or her no previous experience in performing this danger-
food. In mineralocorticoid deficiency, postural ous test in children). The ACTH test is accom-
hypotension or an increase in pulse rate on plished by the administration of 0.25 mg of
standing may be seen; a decrease of blood pres- intravenous (i.v.) synthetic 1–24 ACTH, or in the
sure of 10 mmHg or an increase in pulse rate of low-dose test 1 μg of synthetic 1–24 ACTH, at or
20 is considered significant (autonomic dys- before 8 a.m. and measuring cortisol at 0, 30 and
function is a relatively common cause of pos- 60 min; a value over 18–20 μg/dL is considered
tural hypotension while mineralocorticoid normal. The metyrapone test is used more rarely
deficiency is much more rare). than an ACTH-stimulation test. Metyrapone
Administration of fluid volume and glucose is administration leads to an 11-OH block so that, in
a necessary therapy along with glucocorticoid a normal subject, cortisol decreases while ACTH
replacement. Serum sodium will be low, and and 11-OH deoxycorticosterone increase (as do
serum potassium will be high if mineralocorti- urinary 17-OH corticosteroids), indicating normal
coid secretion is low; replacement of sodium and ACTH reserve. Thus an 8 a.m. serum cortisol,
mineralocorticoid is an appropriate therapy. 11-deoxycortisol, and ACTH are determined, and
246 10 Disorders of the Adrenal Gland

a 24-h urine for 17-OHCS is collected the day tion, so only a few doses per day of replacement
before and the day after the metyrapone is given; glucocorticoids are used. In general, natural
a fourfold increase in 11-deoxycortisol or a 70 % glucocorticoid, cortisol or hydrocortisone, is
increase in urinary 17-OHCS is a normal response. preferable during the growing years, but the more
Metyrapone is given in a dose of 300 mg/m2 every potent, longer lasting glucocorticoids, such as
4 h for a total of six doses over a 24-h period, but prednisone, are used after growth has ceased,
a total dose should be no more than 3 g. mainly because of the ability to cut dosage to
Alternatively, older children are given 30 mg/kg twice per day, or dexamethasone can be used
at midnight (with food), and a blood sample is which can be given once per day. Table 10.1
taken the morning before the metyrapone and the shows the difference in potency of various prepa-
morning after. rations as to glucocorticoid or mineralocorticoid
Other signs of autoimmune phenomena, such effects. Note that prednisone must be converted
as Hashimoto thyroiditis or diabetes mellitus, to prednisolone, and cortisone must be converted
may be found if an autoimmune polyglandular by 11-beta HSD1 to cortisol for biologic activity,
syndrome is suspected. Antibodies to the thyroid, and these medications are therefore given in rela-
the pancreas, or the ovaries may be obtained in tively higher dosage than the more active forms.
such a situation. Prednisone and cortisol should not be used for
emergency treatment because of the lag in time
Treatment of Hypoadrenal States for the conversion to be carried out. The secre-
Note: There is a well-known effect of immunosup- tory rate of cortisol is usually accepted to be
pression caused by glucocorticoids, but in gluco- 6–8 mg/m2 (see discussion above for details).
corticoid deficiency states, it is essential that the Because oral absorption is half the parenteral
glucocorticoids are administered in the recom- absorption, the rate is doubled and administered
mended doses to replace the deficit or to treat a orally three times per day, with most given in the
stressful situation including infections. In these morning, unless the problem is virilizing CAH,
situations, there should be no immunosuppres- when the time of administration may change
sive effect from glucocorticoid administered for (see Chap. 8). Thus for a 1-m2 child, the oral dose
these reasons but if they are not administered to would be 12–14 mg, but because 5- or 10-mg tab-
replace the deficit shock due to acute adrenal lets are the only available oral form, 15 mg is
insufficiency may occur. used, with 5 mg t.i.d. or 10 mg by giving 2.5 mg
If adrenal function is inadequate, the goal is in the morning, 2.5 mg in the early afternoon, and
the replacement of glucocorticoid, or if necessary 5 mg at bedtime. Suspensions made by com-
mineralocorticoid, in physiologic dosage (see pounding pharmacy should not be used for treat-
Chap. 14 for more discussion of acute adrenal ment due to irregularity of the amount of
insufficiency). This is difficult in view of the glucocorticoid in each dose. However, smaller
episodic secretion of cortisol and circadian varia- doses for infants may be carefully prepared at

Table 10.1 Glucocorticoid preparations and relative glucocorticoid and mineralocorticoid effect
Glucocorticoid effect (based Mineralocorticoid effect
on anti-inflammatory effect with [fludrocortisone Time of biologic
cortisol = 1.0) (Florinef) = 1.0] action (h)
Cortisol (hydrocortisone) 1 0.005 8
Cortisone 0.8 0.005 8
Prednisone 4 0 18
Prednisolone 4–5 0 16–36
6α-Methylprednisolone 5–8 0 16–36 est
Dexamethasone 27–66 0 36–54
9α-Fluorocortisone 15 1
Disorders of the Adrenal Cortex 247

home by adding a half of a 5-mg tablet to a given needed to keep the Florinef dose in the 0.05 to
amount of water or formula before each dose and 0.15 mg/day range. Usually older children will
calculating the amount of the suspension to be self-select the correct amount of salt to meet their
administered which will yield the desired dose needs, but infants may need added salt. Sodium
after shaking it to briefly resuspend it. The cor- chloride or table salt is given as measured by the
rect dosage cannot be measured by laboratory teaspoon or by a test tube marked to hold 1 g or
methods, but if the child feels well, vital signs are more. Salt tablets are available which often con-
normal and the child is not gaining excessive tain 1 mg each and a solution might be made in
weight, nor is demonstrating a decreasing growth which the salt tablet or portion of it is dissolved in
rate, the dosage is likely to be appropriate (see water or formula and a calculated amount to
Chap. 8 for dosage in virilizing CAH). In times deliver the required dose be given to the infant.
of stress, fever, surgery, or accidents (but not usu- Note that cortisol and cortisone have some miner-
ally psychological stress), the dose is doubled or alocorticoid effect (Table 10.1), whereas the
tripled until the condition is resolved. At times of more potent glucocorticoids do not; if switching
severe stress (severe accident or shock) or a sur- from one preparation to the other, watch for a
gical procedure when the child is not able to take change in sodium balance or adjust/add miner-
medication orally, injectable glucocorticoid in alocorticoid to the regimen. Shock is a real pos-
the form of Solu-Cortef in a Redivial© that com- sibility if mineralocorticoid deficiency or sodium
bines diluent and powdered medication in one deficiency develops.
action of a rubber stopper is easiest to use; gener- It is imperative that if hypothyroidism and glu-
alized doses are 25 mg for infants, 50 mg for cocorticoid deficiency coexist, the glucocorticoid
smaller children, and 100 mg for adolescents to be instituted before thyroid hormone, or the mini-
be given immediately upon noting the severe mal glucocorticoid that may be present endoge-
condition and certainly before the child is taken nously will be more rapidly metabolized, leading
to the emergency department (ED) at times of to acute glucocorticoid-deficient crisis.
emergency. No danger occurs in giving one dose
of the medication inappropriately, but not giving Glucocorticoid Withdrawal
the dose when needed can be serious or fatal. Iatrogenic glucocorticoid deficiency is caused by
Intramuscular (i.m.) cortisone takes hours to work the abrupt discontinuation of glucocorticoids
and should never be used as emergency treatment. after adrenal suppression. This may occur after
The child should wear a MedAlert bracelet or therapy for a glucocorticoid-responsive disease
necklace stating the steroid dependence and a let- or after the removal of a glucocorticoid-secreting
ter, telling any ED doctor the problem, should be adrenal tumor or condition, such as one causing
carried at all times if possible. In the ED, sugar, Cushing syndrome. Generally, more than
fluid, glucocorticoid, and if salt losing is part of 10–14 days of glucocorticoid therapy will lead to
the problem, NaCl and mineralocorticoid are suppression of endogenous ACTH secretion and,
given immediately. As stated above the letter or with increased use, some degree of atrophy of the
MedAlert bracelet might state Addison’s disease adrenal gland. This process must be reversed
even if the child has salt-losing congenital adrenal before the individual is able to mount a response
hyperplasia for ease of communication. to stress. Depending on the dose, potency of the
When there is salt losing, mineralocorticoid is glucocorticoid, and length of therapy, it may take
given as 9α-fluorohydrocortisone, or Florinef months for ACTH to increase and more months
which allows salt retention. Dosage is meant to for cortisol to follow suit. Nighttime administra-
keep body sodium in an adequate range, so the tion of exogenous glucocorticoid is more likely
plasma renin activity (PRA) is kept in the normal to affect the peak of ACTH secretion and cause
range; Florinef is increased if PRA is too high longer suppression, whereas every-other-day
and decreased if PRA is too low or if blood pres- glucocorticoid administration has less effect.
sure rises too high. Florinef is given once or twice The tolerance of a patient for glucocorticoid
per day. Oral sodium supplementation may be discontinuation is difficult to define, but it is prudent
248 10 Disorders of the Adrenal Gland

to wean the child off slowly rather than cutting Aldosterone Insufficiency
the therapy immediately if any question of adre- or Resistance
nal suppression is seen. Symptoms of the steroid-
withdrawal syndrome that occurs if weaning is The production of aldosterone may be impaired in
too rapid include headache, myalgia, arthralgia, 11-beta hydroxylase deficiency congenital adrenal
and fever, although patients receiving glucocorti- hyperplasia or primary adrenal failure. ACTH defi-
coids for specific diseases may experience resur- ciency will not cause aldosterone deficiency.
gence of symptoms of those diseases and confuse Resistance to aldosterone occurs in the new-
the situation. Many schemas exist for weaning an born demonstrated by elevated plasma aldoste-
individual off glucocorticoids; the following is rone levels and a tendency toward hyperkalemia
only one possibility. The dose of glucocorticoid and hyponatremia which resolve several days
may be decreased 25 % per week until the physi- after birth. Alternatively, resistance may be due
ologic range is reached at which time weaning to pseudohypoaldosteronism (#177735 PSEUDO
slows considerably until the glucocorticoid is HYPOALDOSTERONISM, TYPE I, AUTOSO
discontinued. Normal endogenous production is MAL DOMINANT; PHA1A) due to mutations in
about 6–8 mg/m2/day (see discussion above), but the aldosterone receptor (a member of the nuclear
oral medication is only 50 % absorbed, and so the receptor subfamily 3, group C, member 2 gene
physiologic ranges start at double this secretory (NR3C2)) found in autosomal dominant pattern.
rate. For example, for a child taking 20–30 mg of Clinical features include dehydration although
prednisone divided into two doses per day (which some patients are asymptomatic. Laboratory
is equivalent in bioactivity to about 120–160 mg findings include metabolic acidosis with hypona-
of cortisol) for more than 1 month, the dose may tremia, hyperkalemia, increased serum aldoste-
be cut to 15 mg of prednisone for 1 week, to rone, and increased plasma renin activity.
10 mg for 1 week, to 5 mg for 2 weeks, and then Treatment is accomplished with sodium chlo-
switched to 20 mg of hydrocortisone (divided ride administration.
into two doses per day) for 1 week and 10 mg for A group of five conditions compromise pseudo-
1 week, and then the medication may be given hypoaldosteronism type II; (%145260 ICD+
only in the morning until the a.m. cortisol (mea- PSEUDOHYPOALDOSTERONISM, TYPE IIA;
sured before the cortisol dose) is demonstrated to PHA2A) is due to mutations on chromosome
be greater than 10 μg/dL. With long term sup- 1q31-q42, PHA2B, due to mutations on the WNK4
pression weaning must proceed slower. An a.m. gene on chromosome 17q21, PHA2C due to
serum cortisol greater than 15–20 μg/dL indi- mutations on the WNK1 gene on chromosome
cates adequate basal adrenal function, but the 12p13, PHA2D due to mutations on the KLHL3
subject may not yet have adequate reserves of gene on chromosome 5q31, and PHA2E due to
ACTH for stress. Thus an ACTH-stimulation test mutations in the CUL3 gene on chromosome 2q36.
using 0.25 mg i.m. or i.v. cosyntropin or, a 1 μg Administration of Florinef should cause reso-
intravenous low-dose test, with a measurement of lution of hyponatremia and hyperkalemia in
serum cortisol in 60 min greater than 20 μg/dL aldosterone deficiency but not in aldosterone
indicates good reserve. A stress dosage of gluco- resistance. Administration of sodium chloride as
corticoids (2–3 times normal basal secretion of used in salt-losing congenital adrenal hyperplasia
8 mg/m2/day given parenterally if the child can- will help resolve the electrolyte abnormalities in
not swallow or is vomiting or orally at double this pseudohypoaldosteronism.
dose) must be administered if the child has an ill-
ness or accident or if surgery is required before
adrenal reserve returns (see Chap. 8 concerning Adrenal Calcifications
CAH). It is useful for the child to wear a MedAlert
bracelet to notify the emergency staff of reduced Adrenal calcifications may be asymptomatic or
adrenal reserve should an emergency room visit may be associated with pathological states.
be required. Thus Addison’s disease; adrenal hemorrhage;
Disorders of the Adrenal Cortex 249

Waterhouse-Friderichsen syndrome; tumors described earlier for Addison’s disease. Delay in


including neuroblastoma, pheochromocytoma, diagnosis is frequent with subtle symptoms preced-
adrenal adenoma, or carcinoma; and infections ing the diagnosis by years.
including TB and Wolman disease may have
associated adrenal calcification. Cushing Disease: Hypercortisolism
due to Increased ACTH Secretion
(Fig. 10.5)
Hyperadrenal States An abnormal regulation of the pituitary–adrenal
axis leads to the specific diagnosis of Cushing
Cushing Syndrome disease (called bilateral adrenal hyperplasia,
Cushing syndrome refers to the general class of because of the enlarged appearance of both adre-
disorders with increased glucocorticoid effect, nal glands, even though the etiology is in the
including those due to exogenous glucocorticoid hypothalamic–pituitary axis; this should not be
intake. The specific causes of endogenous hyper- confused with congenital adrenal hyperplasia).
cortisolism include conditions secondary to The original report by Cushing describes a patient
increased pituitary ACTH secretion (only this with a basophilic adenoma and physical signs of
form is termed Cushing disease), ectopic ACTH hypercortisolism, but it was not until 50 years
secretion, autonomous cortisol secretion by the later that the primary importance of the pituitary
adrenal gland, or glucocorticoid therapy be it microadenomas in these patients was proven.
oral, parenteral, inhaled, or dermal. Before age The fact that about 20 % of patients appropriately
7 years, the usual cause of Cushing syndrome is treated with pituitary microadenomectomy have
an adrenal tumor, whereas thereafter, increased recurrences indicates that basic hypersecretion of
pituitary ACTH secretion characteristic of hypothalamic CRF may be the etiologic agent for
Cushing disease becomes a more common etiol- a minority of patients with Cushing disease.
ogy. Hypercortisolism has wide-ranging effects Cushing disease in childhood first manifests
on the body, including decreased growth rate; with growth failure and weight gain in the truncal or
weight gain in a general truncal or centripetal centripetal pattern, with thin and weak extremities.
pattern, leading to the characteristic buffalo hump Other features may include the classic cushingoid
of excessive adipose deposition on the back and a appearance of a buffalo hump of adipose tissue on
characteristic round face (moon facies); muscle the back of the neck; purple striae of the trunk due
weakness, especially in the distal regions, lead- to thinning of the skin and exposure of the capillar-
ing to thinning of the extremities while weight ies; sometimes, but not invariably, pigmentation of
gain progresses; thinning of the skin, leading to the flexural surfaces and gums; lack of energy; and
purple striae (or the so-called cigarette paper skin the early appearance of acne or pubic hair due to
referring to the extreme thinness of the skin that increased DHEA secretion because of increased
leads to the visibility of the underlying capillar- ACTH stimulation. Excess glucocorticoids cause
ies); hypertension; fatigue; and insulin resistance. protein catabolism and results in muscle wasting
Osteopenia or osteoporosis can lead to multiple and weakness. The personality of patients with
fractures of the spine and other bones. Acne and Cushing disease may be abnormal, and some
hirsutism as well as delayed puberty are often describe the children as obsessive. The diastolic and
found in older children. Because the mineralo- systolic blood pressure may be elevated.
corticoid receptors respond to cortisol especially Because of the small size of the usual lesion,
in severely elevated levels, an excess of cortisol usually less than 10 mm, neurologic symptoms or
leads to salt retention and hypertension (although signs are not found in Cushing disease unless the
such activity is usually minimized by the mass has progressed markedly. Contrast-enhanced
11β-hydroxylation by 11-beta HSD2 of excess MRI in some cases may indicate the location of
cortisol to cortisone, which is inactive). If the the microadenoma but is not always success-
cause of the excess glucocorticoid is excess ful. Indeed, unless the surgeon has great experi-
ACTH, hyperpigmentation will be found, as ence in individual detection of the microadenoma
250 10 Disorders of the Adrenal Gland

Third ventricle Hypothalamus


Neuroendocrine
cell nuclei

CRF-secreting cells

Superior
hypophyseal
artery
Stalk

Long portal
vessels
Inferior
Adrenocorticotrophs hypophyseal
artery

Secreting cells

Anterior Posterior
pituitary pituitary

Hormone Short portal


secretion vessel
Glomerulosa
+ ACTH
Fasciculata

Reticularis

Medulla

Aldosterone Cortisol Androgens Catecholamines

Fig. 10.5 Endocrine changes of Cushing disease due to values in the presence of elevated cortisol values. ACTH-
increased ACTH secretion escaping from feedback inhibi- secreting pituitary adenomas or ectopic production of
tion leading to inappropriately normal or elevated ACTH ACTH will present in a similar manner

of Cushing disease and the subtle changes in Microadenomectomy may have to be repeated
appearance between the microadenoma and nor- and may ultimately fail in some patients.
mal pituitary tissue may be missed. The majority Hypophysectomy may be required leading to
of patients successfully treated with removal of hypopituitarism, but these deficiencies can be
the microadenoma of the pituitary gland ulti- treated. Irradiation of the pituitary gland is an
mately regain normal diurnal variation of cortisol alternative approach and may also lead to hypopi-
and ACTH and the growth hormone deficiency tuitarism in subsequent years.
that may accompany the microadenoma usually Medical treatments are available. Metyrapone
resolves although it may take months to years. will lead to only a temporary suppression of adrenal
Disorders of the Adrenal Cortex 251

production of glucocorticoids while ortho,para- pigmentation is known as Nelson syndrome which


DDD (mitotane) leads to a more permanent sup- is more often found in adults than children.
pression of adrenal function although substantial
side effects limit its usefulness in children. If the Cushing Syndrome due to Autonomous
adrenal glands are removed in an attempt to treat the Cortisol Secretion (Fig. 10.6)
disease without successful removal of the etiology
which is the increase in ACTH secretion, enlarge- Nodular Adrenal Hyperplasia or Adrenal
ment of the unsuppressed pituitary gland leads to Tumors
pressure effects on the surrounding tissue, including Under the age of 7 years adrenal tumors take
the optic chiasm; this in addition to extreme hyper- precedence in the etiology of Cushing syndrome

Third ventricle Hypothalamus


Neuroendocrine
cell nuclei

CRF-secreting cells

Superior
hypophyseal
artery
Stalk

Long portal
vessels
Inferior
Adrenocorticotrophs hypophyseal
artery

Secreting cells

Anterior Posterior
pituitary pituitary

Hormone Short portal


secretion vessel
Glomerulosa
ACTH
Fasciculata

Reticularis

Medulla

Aldosterone Cortisol Androgens Catecholamines

Fig. 10.6 Endocrine changes of Cushing syndrome due to autonomous production of cortisol. Elevated cortisol secretion
exerts negative feedback inhibition on the hypothalamic pituitary axis leading to low ACTH values
252 10 Disorders of the Adrenal Gland

while over 7 years Cushing disease is more There are two micronodular types of nodular
frequent. adrenal hyperplasia, primary pigmented nodular
Autonomous cortisol secretion may be seen in adrenocortical disease (PPNAD) or isolated
adrenal carcinoma or in single or multiple autono- micronodular adrenocortical disease. PPNAD
mous nodules of the adrenal gland(s). Adenomas (10489 PIGMENTED NODULAR
or carcinomas of the adrenal glands may be found ADRENOCORTICAL DISEASE, PRIMARY, 1;
in conditions of asymmetry such as the Beckwith– PPNAD) is characterized histologically by pig-
Wiedemann syndrome (see Chap. 12). A carci- mented nodules in the adrenal cortex which
noma of the adrenal gland often is difficult to secrete glucocorticoids and androgens; these
differentiate from a benign tumor of the gland; tumors remarkably increase glucocorticoid
pathologic criteria are available, but often, clini- secretion in response to a high-dose
cal observation is necessary to define the differ- dexamethasone-suppression test while in the nor-
ence between a carcinoma and an adenoma. mal situation dexamethasone administration
Carcinomas of the adrenal gland may have defects should suppress glucocorticoid secretion. The
in 3β-hydroxylase and secrete large amounts of clinical findings of Cushing syndrome will
DHEA as well as cortisol, leading to a greater develop. PPAND is usually found as part of the
amount of virilization than found with the other Carney complex (#160980 CARNEY COMPLEX,
causes of Cushing syndrome such as adrenal ade- TYPE 1; CNC10). The Carney complex is an
nomas which tend to produce only glucocorti- autosomal dominant multiple endocrine neopla-
coids. Testosterone may be produced directly by sia condition with many physical manifestations
such tumors, and severe virilization has been of the eye (conjunctival and scleral pigmenta-
seen; the differential diagnosis should include vir- tion), the heart (atrial or ventricular myxoma
ilizing ovarian tumors in girls. Because the natu- with congenital heart failure), the skin (numerous
ral history of any condition will start with subtle pigmented nevi, sometimes blue, and lentigines),
signs, apparent premature adrenarche without hirsutism and red hair, and a tendency to a host of
other definitive signs of virilization may be the neoplasias including subcutaneous tumors, Sertoli
first indication of the presence of an adrenal tumor cell tumors, mammary ductal fibroadenomas,
and demonstrates the importance of follow-up Schwannomas, thyroid carcinomas, and pheo-
even in apparently benign conditions. Feminizing chromocytomas. Pituitary adenomas will usually
tumors of the adrenal gland have been rarely secrete growth hormone and can cause gigantism
reported in childhood. The treatment of adrenal in younger individual or acromegaly in older indi-
tumors is surgical removal although suppression viduals while prolactin secretion and galactorrhea
of the steroid production with the ortho,para- may also occur. Differentiated thyroid carcinoma
DDD (mitotane) has been used when surgery was is associated with the Carney complex as are cal-
ineffective in removing all traces of the tumor. cified Sertoli cell tumors of the Leydig cells which
Nodular adrenal hyperplasia is characterized may be indicated by the development of gyneco-
by areas of active autonomously secreting adre- mastia. The Carney complex is found with muta-
nal tissue in an ACTH-independent manner inter- tions in the cAMP-dependent protein kinase,
spersed with areas of quiescent gland on regulatory, type I, alpha gene.
histologic examination which is due to feedback
inhibition of ACTH secretion by the secretion of Ectopic ACTH Secretion
cortisol from the active nodules. The macronodu- Ectopic secretion of ACTH is exceedingly rare in
lar form may be associated with McCune– childhood but has been reported in Wilms tumors
Albright syndrome (see Chap. 9) in association and in islet cell carcinoma of the pancreas. Serum
with other hyperactive endocrine conditions, ACTH values may exceed 1,000 pg/mL, although
polyostotic fibrous dysplasia, and café au lait modest elevations may be seen at first. The excep-
spots of the skin or may be found in multiple tional elevations can stimulate mineralocorticoid
endocrine neoplasia type I (MEN 1). production, although mineralocorticoids are not
Disorders of the Adrenal Cortex 253

Poor growth with weight gain

Evaluate as described in Chapter 4 for hypothyroidism, Chapter 5 for growth hormone deficiency, Chapter 7 for pseudohypoparathyroidism

Exogenous source of glucocorticoids by mouth, skin or respiratory system


No Yes

Obtain 24 hour urinary free cortisol or Overnight dexamethasone suppression test

AM cortisol not suppressed AM cortisol suppressed

Elevated Normal two times Exogenous Cushing syndrome

Serum ACTH Cushing disease/syndrome unlikely


common obesity possible

Extremely high High or AM range values Low


found in the PM

Perform high dose Adrenal adenoma or


dexamethasone suppression test carcinoma possible

Not suppressed Suppressed

Exotopic ACTH Cushing disease possible


secretion possible

Fig. 10.7 The evaluation of hypercortisolism

normally very responsive to ACTH in the physio- differential diagnosis is often between exogenous
logic state, leading to salt retention and potassium obesity and Cushing disease (Fig. 10.7).
excretion; hypertension, hypokalemic alkalosis, Urinary metabolites of cortisol are measured as
and subsequent muscle weakness will result. It is 17-hydroxycorticosteroids (17-OHCS) which
rare to see such excess mineralocorticoid produc- measure both cortisol and its urinary metabolites
tion in Cushing disease. and the more specific urinary free cortisol measur-
ing only free cortisol. Normal urinary free cortisol
Differential Diagnosis of Cushing Disease is 11 ± 5 mg/m2/day. The urinary 17-OHCs and
Serum ACTH is not necessarily strikingly ele- urinary free cortisol may be elevated for age in
vated in Cushing disease; the finding of a concen- exogenous obesity because of the changes in glu-
tration of ACTH more characteristic of the cocorticoid metabolism brought about by obesity,
morning (higher) in the afternoon (i.e., ACTH thereby confusing the diagnosis in an obese indi-
concentration is too high for the time of day) vidual, but in Cushing disease, the values will be
associated with a value of cortisol also too high higher when corrected for body surface area than
for the afternoon or evening is suggestive of those found in obesity (24-h urinary 17-OHCs
abnormality in the regulation of the ACTH– greater than 4.5 mg/m2 and urinary free cortisol
cortisol axis (Fig. 10.3). It must be remembered, greater than 70 g/m2 are highly suggestive of
however, that stress or even depression will ele- hypercortisolism). The great variations in urinary
vate both ACTH and cortisol levels, will be asso- values from day to day and the difficulty in col-
ciated with aberrant diurnal rhythms, and can lecting a full 24-h sample of urine in childhood
falsely suggest Cushing disease. Pseudocushing mandate that at least two baseline urine collections
syndrome denotes a situation with many or most are evaluated and collected for glucocorticoid
symptoms and signs of Cushing disease, but they determinations. The collection should be standard-
are secondary to other conditions including ized by creatinine determination.
severe emotional or pathophysiological stress, A midnight blood sample for cortisol in a nor-
depression, panic disorders, and psychosis as mal individual should reveal a value lower than
well as alcoholism or alcohol withdrawal. As 2 μg/L in the resting individuals in whom an IV
most patients with Cushing disease are obese, the catheter has already been placed so as not to
254 10 Disorders of the Adrenal Gland

disturb the patient; if the patient awakens, corti- following the low-dose test, thereby eliminating
sol will rise and the diagnosis will still be uncer- the need for 48 h of baseline urine collection
tain. An overnight dexamethasone-suppression once again. The high-dose dexamethasone-
test may be used to screen patients for Cushing suppression test is performed to differentiate
disease, but this test has a high incidence of between Cushing disease and Cushing syndrome
false-positive results, although the rate of false from other causes. In the high-dose dexameth-
negatives is low; 1 mg of dexamethasone asone-suppression test, 3.75 mg/m2/24 h or
(0.3 mg/m2 or 15 μg/kg body weight is used for 120 μg/kg/dose of dexamethasone (maximum 2
smaller children) is given orally at midnight, and mg/dose) is given as four doses a day for 2 days,
a serum sample is analyzed for cortisol at 8:00– and urinary 17-OHC excretion on the second day
9:00 a.m. the next morning. In presumptive is compared with the two baseline collections; in
Cushing disease, plasma cortisol will not be sup- Cushing disease, complete suppression of uri-
pressed below 1.8 mg/dL, whereas in exogenous nary 17-OHCs (<1 mg/kg creatinine) and free
obesity, the values decrease below 1.8 mg/dL cortisol will occur, whereas in the ectopic ACTH
which is a negative test. If the results are nega- syndrome or with autonomous cortisol secretion,
tive, Cushing disease is unlikely, but if the results no suppression will be seen over baseline levels.
are positive, the following tests are considered. Remarkably in PPNAD there may be a paradoxi-
Salivary cortisol may replace plasma cortisol cal increase in cortisol secretion after high-dose
measurements in this test. suppression as noted above.
The classic tests for the differential diagnosis Ovine CRF infusion is reported as a useful
of Cushing syndrome are the classic dexametha- measure for the differential diagnosis between
sone-suppression tests. In any test requiring Cushing syndrome and Cushing disease as a cor-
urinary collections, two baseline 24-h urinary tisol value >1.4 μg/dL following a low dose
collections are required and the collection is best dexamethasone suppression test as described
carried out in a clinical research unit if possible above indicates the diagnosis of Cushing syn-
due to the vagaries of collection at home. To drome and should lead to further evaluation.
insure complete collection, results are character- However, severe obesity alters results and may
istically expressed per gram creatinine. The limit the utility of this test. A patient with auton-
low-dose dexamethasone-suppression test uses omous cortisol secretion will have low basal
1.25 mg/m2/24 h or 30 μg/kg/dose up to 0.5 mg per ACTH values, a value over 29 pg/mL leads to
dose given as four doses per day for 2 days; if the the diagnosis of ACTH dependent Cushing syn-
urinary 17-OHCs on the second day of the test drome. A patient with ACTH dependent Cushing
decrease 50 % from the two baseline collections syndrome will not have an increase in ACTH
(or if the 17-OHCs are less than 1 mg/m2 and the with CRF administration. A patient with ectopic
urine free cortisol less than 25 μg/m2), the patient ACTH secretion will have a high basal ACTH
is likely to have exogenous obesity because concentration and will not have an increase in
they are easily suppressible. It is useful to collect ACTH with CRF, whereas a patient with Cushing
serum ACTH and cortisol concentrations early in disease will have a large increase in ACTH,
the morning (6–8 a.m.) and at night (best at mid- demonstrating the CRF-responsive nature of the
night but could be as early as 8 p.m.) during the microadenoma.
baseline and dexamethasone-suppression phases Salivary cortisol reflects the free cortisol
of the test to see if serum values also are affected moiety in plasma and is becoming more popular
(e.g., if the cortisol is suppressed to less than in the evaluation of cortisol secretion in chil-
1.8 μg/dL and the ACTH to less than 25 pg/mL); dren. Samples can be collected by spit or by
triplicate sampling is recommended for the swabs. Results can be affected by oral intake,
ACTH and cortisol samples because of pulsatile oral bleeding, tooth brushing, and other daily
secretion. Since it will take a while to receive the activities, so the patient must be carefully pre-
results of a low-dose test, the high-dose dexa- pared for the collection. This salivary method
methasone test is usually carried out immediately has generally been used for late-night collections
Disorders of the Adrenal Cortex 255

or overnight dexamethasone-suppression test- formed if necessary. In the right hands, this proce-
ing. Duplicate samples are suggested to increase dure is the treatment of choice. Removal of the
accuracy. basophil adenoma secreting ACTH usually leaves
Invasive sampling procedures more often are the remaining quiescent basophils which do not
applied in adult patients than in children; if the secrete ACTH. It may take months, up to 18, for the
technical expertise is available, however, these basophils to recover normal ACTH secretion, and
techniques may be applied in pediatrics. Adrenal the secondary adrenal insufficiency must be treated
venous sampling for cortisol or other adrenal with glucocorticoid replacement therapy until then.
gland metabolites is difficult in children, but this Previous studies demonstrated the utility of
technique may be used in older teenagers to further o,p′-DDD, an adrenolytic agent, to diminish
evaluate patients suspected to have an adrenal adrenal function, but other methods have sup-
etiology of Cushing syndrome and can indicate planted its use in all but inoperable patients.
whether a unilateral adrenal tumor is responsible Metyrapone, an 11-hydroxylase inhibitor, has
for the cortisol secretion. For the evaluation of been used with some temporary success, but
Cushing disease, petrosal sinus sampling for ACTH levels will soon increase and overwhelm
ACTH may indicate the side and location of a the block in cortisol production, leading to
pituitary or extrapituitary source for the ACTH to renewed symptoms of hypercortisolism.
narrow the surgical approach necessary for treat- Mifepristone is a glucocorticoid receptor antago-
ment. These highly technical procedures should nist which is used for inoperable Cushing syn-
only be carried out by experienced hands. drome but it is not approved for pediatric use.
Noninvasive radiologic procedures meet with Radiotherapy has been used in some series, but
mixed success, especially in the CNS. CT scan- the risk of hypopituitarism developing after
ning or MRI with gadolinium of the pituitary radiotherapy has led most clinicians to use other
region may not be helpful because of the small methods of treatment. Bilateral adrenalectomy
size of some microadenomas, and the presence of will cure the hypercortisolism but leave the pitu-
silent adenomas in many normal individuals may itary tumor. After a period, the hypersecretory
confuse the issue. The adrenal glands may both pituitary tumor, released from the suppressive
appear enlarged on abdominal CT scans in effects of hypercortisolism, may grow and secrete
Cushing disease, whereas a solitary nodule or increasing amounts of ACTH: hyperpigmenta-
tumor or nodular adrenal hyperplasia may be tion and an enlarging pituitary tumor of Nelson
identified on high-quality CT scans or MRI. The syndrome result. Of course, if the adrenal glands
presence of an abnormality on imaging may be are removed, both glucocorticoids and mineralo-
helpful in pointing to a disorder, but imaging is corticoids will have to be replaced for the life-
not an acceptable way to eliminate a disorder. span of the patient.
Newer techniques of imaging such as fluorine-18 The aim of treatment of autonomous adrenal
fluorodeoxyglucose PET combined with CT may tumors and ectopic ACTH-secreting tumors is to
add useful information. Exploratory laparotomy eradicate the offending neoplasm. If the ectopic
may yet be required to localize the tumor. ACTH-secreting tumor is inoperable, an adrenal
blocking agent or bilateral adrenalectomy is used
Treatment of Cushing Syndrome to control the hypercortisolism.
If the diagnosis of Cushing disease is established, The goal of medical treatment of Cushing syn-
the treatment may be medical, radiotherapy, or sur- drome after the correction of the hypercortisolism
gery. Transsphenoidal microadenomectomy is a is the replacement of glucocorticoids in appropri-
useful method of curing the patient of the microad- ate dose to mimic normal adrenal secretion and to
enoma that causes the problem and leaving the rest avoid the suppression of statural growth. In the
of the pituitary gland intact. Experience with this months after the removal of the microadenoma
particular tumor is essential if the surgeon is going of Cushing disease, the remaining suppressed
to remove the appropriate part of the gland. corticotrophs will not function, and replacement
Recurrence is possible, but reoperation can be per- glucocorticoid therapy will be required. The initial
256 10 Disorders of the Adrenal Gland

dose of cortisol will have to be relatively high, or jects when standing for 4 h but decreases in adre-
the patient may experience the malaise and leth- nal adenomas since this condition is usually not
argy characteristic of glucocorticoid withdrawal. controlled by the stimulation of angiotensin as in
If adequate cortisol is not supplied after surgery, the normal situation; however, some adenomas
the child might also go into the shock of acute are indeed responsive to angiotensins, so the test
adrenal insufficiency. Although the desired dose is not completely reliable. Selective venous sam-
of hydrocortisone is approximately 6–8 mg/m2 pling will indicate unilateral or bilateral disease,
parenterally or double that dose orally, the initial but the technique is quite difficult especially in
dose may have to be twice or more for several children. Imaging by an adrenal iodocholesterol
weeks, with gradual weaning to the lower dose as (NP-59) scan with dexamethasone suppression or
necessary (see earlier). After 9–18 months, normal CT and MRI has been useful in localizing the
adrenal function will return in the majority of lesion. Treatment is sometimes accomplished by
cases of Cushing disease treated with transsphe- high doses of spironolactone but usually by adre-
noidal microadenomectomy. Adrenal suppression nal surgery especially if the disease is unilateral.
is also described above. It is essential to eliminate secondary hyperaldo-
A recent study evaluated growth after treat- steronism before initiating treatment. Aldosterone
ment of Cushing disease versus autonomous will rise when blood volume is depleted due to
glucocorticoid-secreting adrenal nodules. The fluid loss or blood loss, sodium is depleted, or
results showed greater growth after the removal of potassium is elevated whether due to disease or
nodules than the removal of the pituitary microad- with the treatment of a condition by medication.
enomas of Cushing disease partially because Familial hyperaldosteronism (*605635
Cushing disease occurs in older children, partially HYPERALDOSTERONISM, FAMILIAL, TYPE
because the androgen secretion which may II) is due to an adrenal adenoma, bilateral adrenal
accompany Cushing disease is usually absent in hyperplasia, or both. It is not treatable with dexa-
autonomous glucocorticoid-secreting nodules, methasone as are the following conditions.
and partially because of the higher prevalence of
vertebral fractures in Cushing disease due to Hypertensive Conditions Treated
osteopenia. While the removal of microadenomas with Glucocorticoids
of the pituitary is usually quite successful, growth Glucocorticoid-suppressible hyperaldosteronism
hormone deficiency can occur and further add to (#103900 #103900 ICD+ GLUCOCORTICOID-
the discrepancy in growth between the two types REMEDIABLE ALDOSTERONISM; GRA
of conditions. or HYPERALDOSTERONISM, FAMILIAL,
TYPE I at 8q21) is a rare autosomal dominant
condition due to fusion of the cytochrome P450,
Hypertension due to Adrenal Disease subfamily XIB, polypeptide 1 gene (CYP11B1,
610613), and the cytochrome P450, subfamily
Primary Hyperaldosteronism XIB, polypeptide 2 gene (CYP11B2, 124080)
Primary hyperaldosteronism (or Conn syndrome) leading to elevated aldosterone production; the
is usually unilateral. With this condition serum or increased production of aldosterone is not
urinary aldosterone is elevated, plasma renin caused by a plasma renin activity as PRA levels
activity is low, and potassium is elevated with are suppressed. There is an associated increase
hypokalemic alkalosis, hypertension (due to in 18-oxocortisol and 18-hydroxycortisol.
sodium retention), and polyuria. Aldosterone is Hypertension with low potassium results, but this
elevated due to autonomous production from the condition is treated with glucocorticoid adminis-
adrenal gland rather than by stimulation by tration. 17α-Hydroxylase deficiency (*202110
angiotensin II and PRA is suppressed. Plasma ADRENAL HYPERPLASIA, CONGENITAL,
aldosterone concentration rises in normal sub- DUE TO 17α-HYDROXYLASE DEFICIENCY)
Adrenal Medulla 257

and 11β-hydroxylase deficiency (*202010 Bartter Syndrome


ADRENAL HYPERPLASIA, CONGENITAL, Bartter syndrome refers to several autosomal
DUE TO 11β-HYDROXYLASE DEFICIENCY) recessive disorders characterized by decreased
congenital adrenal hyperplasia both produce a sodium chloride reabsorption in the thick ascend-
hypertensive state (see earlier in Chap. 8). ing loop of Henle which leads to salt wasting,
11-Hydroxysteroid dehydrogenase deficiency hypokalemia, hypochloremic metabolic alkalo-
interrupts the normal conversion of cortisol to sis, and hypercalciuria with increased plasma
cortisone by 11-hydroxysteroid dehydrogenase renin activity and hyperactivity of the renin–
(218030 CORTISOL 11β-KETOREDUCTASE angiotensin system with increased plasma aldo-
DEFICIENCY at 16q22) and leads to apparent sterone; hypomagnesuria is often found as well.
mineralocorticoid excess caused by the excess There are antenatal severe forms of Bartter syn-
mineralocorticoid effect exerted by cortisol. drome (#601678 ICD+ BARTTER SYNDROME,
The condition may be noted at birth and require ANTENATAL, TYPE 1) (#241200 ICD+
glucocorticoid administration to decrease cortisol BARTTER SYNDROME, ANTENATAL, TYPE
production and mineralocorticoid antagonism by 2) as well as forms (#602522 BARTTER
the use of spironolactone. Alternatively, the con- SYNDROME, TYPE 4A) (#613090 BARTTER
dition may be an acquired defect due to licorice SYNDROME, TYPE 4B) associated with senso-
ingestion (natural licorice such as found in rineural deafness.
Europe, not the red candy ropes more prevalent
in the USA); the treatment is to discontinue lico- Gitelman’s Syndrome
rice. Natural licorice as found in Europe, Asia, Gitelman’s syndrome (#263800 ICD+
and Africa exerts both mineralocorticoid proper- GITELMAN SYNDROME) is an autosomal
ties and glucocorticoid properties by inactivating recessive disorder due to mutation in the thiazide-
11-beta-hydroxysteroid dehydrogenase (type 2) sensitive NaCl cotransporter (SLC12A3). This
which would ordinarily convert active cortisol to condition is associated with renal potassium and
inactive cortisone, thereby leading to increased magnesium wasting with increased plasma renin
cortisol effect. activity resulting in polydipsia, hypomagnese-
mia, hypocalciuria, and hypokalemic alkalosis.
Familial Glucocorticoid Resistance Patients may demonstrate generalized muscle
Familial glucocorticoid resistance (#615962 weakness, muscle cramps, tetany, seizures, par-
GLUCOCORTICOID RESISTANCE, GENERA- esthesias, and episodic paralysis after strenuous
LIZED) is an autosomal dominant condition due exercise.
to abnormality of the glucocorticoid receptor
caused by a mutation in the nuclear receptor sub-
family 3, group C, member 1 gene (NR3C1). Adrenal Medulla
Glucocorticoid values are quite high, but these
levels do not cause findings of excessive gluco- Normal Physiology
corticoids such as found in Cushing syndrome.
Since glucocorticoids cannot exert feedback to The adrenal medulla is derived from neuroecto-
suppress ACTH secretion due to glucocorticoid derm and produces catecholamines in the chro-
resistance, patients have increased products of the maffin cells. Chromaffin cells also are located in
adrenal gland and the other steroids have working the sympathetic ganglion and the organ of
receptors. Thus findings of hypermineralocorti- Zuckerkandl (anterior to the aorta), and although
coidism and virilization may result. Treatment is the extraadrenal chromaffin cells mostly regress
accomplished by increased doses of dexametha- in the postnatal period, they remain a potential
sone and if necessary treatment of the hyperten- site of tumor formation and must be considered
sion with spironolactone. in the diagnostic process.
258 10 Disorders of the Adrenal Gland

The catecholamine biosynthetic pathway pro- tern (#171300 PHEOCHROMOCYTOMA at


ceeds from L-tyrosine to L-Dopa (via tyrosine 11q23, 1p). They are a component of the multiple
hydroxylase) to dopamine (via aromatic-L-amino endocrine neoplasia syndromes (see below), neu-
acid decarboxylase) to norepinephrine (via dopa- rofibromatosis (*162200 NEUROFIBROM
mine β-hydroxylase) to epinephrine [via phenyl- ATOSIS, TYPE I; NF1 at 17q11.2), and von
ethanolamine-N-methyltransferase (PMNT)]. Hippel–Lindau disease (*193300 VON HIPPEL-
The last step occurs only in a specific sympa- LANDAU SYNDROME; VHL at 3p26-p25) and
thetic nervous tissue, the adrenal medulla; the may develop before or after other components of
high concentration of cortisol in the blood supply the syndromes are noted.
to the adrenal medulla induces PMNT and Characteristic signs and symptoms of pheo-
accounts for the transformation of significant chromocytomas include hypertension (more
amounts of norepinephrine into epinephrine in often constant than episodic in children com-
this location but not in other chromaffin tissues. pared with adults), weight loss, headache, vom-
Conversely in primary adrenal failure when there iting, and, more rarely in childhood than in
is no high concentration of cortisol reaching the adults, paroxysmal episodes of tachycardia,
blood supply of the medulla, epinephrine produc- flushing, sweating, palpitations, or anxiety.
tion decreases. Since adrenal failure can lead to Postural hypotension due to volume depletion
hypoglycemia and since it is the catecholamines may occur. Diagnosis is made by the demonstra-
of the adrenal medulla that produce the adrener- tion of elevated plasma or 24-h urinary fraction-
gic symptoms of hypoglycemia, a person may ated metanephrines, which include metanephrine
experience low blood sugar without realizing it and normetanephrine, about fourfold higher than
as a form of hypoglycemia unresponsiveness. the upper limits of normal range. Various drugs
Catecholamines are metabolized by monoamine interfere with this testing including acetamino-
oxidase (MAO) and catechol-O-methyltransferase phen, tricyclic antidepressants, phenoxybenza-
(COMT). Vanillylmandelic acid (VMA) and mine, and decongestants. HVA and VMA
homovanillic acid (HVA) are deaminated metab- excretion in spot urine collection remain helpful
olites of catecholamines and are measured when in the diagnosis of neuroblastoma but have been
excessive catecholamine production is suspected, replaced in the diagnosis of pheochromocytoma
or where metabolism of catecholamines also is or paragangliomas by fractionated urine or
high, such as in neuroblastoma. Epinephrine and plasma catecholamines. Supine measurements of
norepinephrine mediate their actions through plasma metanephrines are helpful if pheochro-
alpha and beta receptors, while dopamine medi- mocytoma is suspected, but standing measure-
ates its action through a family of “D receptors” ments are higher normally; if the sample was
such as the D2 receptors in the lactotrophs which collected in a previously inserted intravenous
when activated suppress prolactin secretion. line, stress will not affect the value nor lead to a
false positive. Plasma catecholamine concentra-
tions may not add any information to the urinary
Disorders of the Adrenal Medulla collections, which serve as a reflection of the
integrated catecholamine production over the
Pheochromocytoma previous day. Tumors secreting norepinephrine
Pheochromocytomas are derived from the chro- and normetanephrine are more likely associated
maffin cells of the adrenal medulla; tumors of the with paragangliomas while tumors secreting epi-
extraadrenal chromaffin cells are considered as nephrine or normetanephrine are more likely to
paragangliomas. Pheochromocytomas are rare in be pheochromocytomas. Provocative pharmaco-
childhood but represent a curable cause of hyper- logic tests are no longer used in the diagnosis of
tension and must be considered in the differential pheochromocytoma or paragangliomas because
diagnosis of high blood pressure. Familial cases of the danger of precipitating a hypertensive cri-
may be inherited in an autosomal dominant pat- sis and because of the specificity of the plasma
Adrenal Medulla 259

and urine collections. Dopamine-secreting Multiple Endocrine Neoplasia


tumors are quite rare, but in the clinical situation
where biochemical indicators of pheochromocy- Multiple Endocrine Neoplasia Syndromes
toma or paragangliomas are negative but one of Multiple endocrine neoplasia syndrome type 1
these two tumors is suspected, determination of (# 131100. MULTIPLE ENDOCRINE
dopamine and HVA can be helpful. NEOPLASIA, TYPE I; MEN1; caused by muta-
An adrenal pheochromocytoma may be tion in the MENEN gene at 11q13) sometimes
located with abdominal CT scanning or with called Wermer syndrome occurs in a dominant
MRI. To ascertain function, scintiscanning with pattern although 10 % of patients have new muta-
the synthetic catecholamine precursor, 131I-meta- tions. MEN type I comprises hyperparathyroid-
iodobenzylguanidine, which is concentrated by ism (tumors are often found in all four parathyroid
chromaffin tissue, and single-photon emission glands and the parathyroid hormone (PTH) will
computed tomography (SPECT)/CT are used. If be inappropriately elevated in view of the
imaging techniques fail, selective venous cathe- increased serum calcium), pituitary adenomas
terization and analysis of catecholamine efflux (increased GH secretion causes elevated serum
may narrow the search for the tumor but only in insulin-like growth factor (IGF)-1 and acromegaly;
older teenagers as such sampling is virtually increased prolactin causes galactorrhea), pancre-
impossible in small children. atic islet cell tumors (hypoglycemia due to insu-
Surgery to remove the tumor is the treatment lin from the beta cells), and duodenal tumors
of choice for pheochromocytoma, but the patient ulcers caused by (gastrin from the alpha cells of
must be prepared for surgery and for invasive the pancreas or the G cells of the stomach or duo-
radiologic studies by adrenergic blockade. denum) as well as skin, adrenocortical, carcinoid,
Medical management and surgical treatment of and central nervous system tumors. Thus hyper-
these conditions are fraught with risk and only an calcemia, hyperprolactinemia, GH excess, and
experienced team should consider approaching a hypoglycemia may result. Usually pheochromo-
child affected by these tumors. The following is a cytomas are not included in MEN type 1, but
summary of the very complex process. Alpha- families rarely are reported with only pheochro-
adrenergic block should be administered before mocytoma and islet cell pancreatic tumors found
β-adrenergic blockade. Phenoxybenzamine is in a complex. This disorder usually is rare in chil-
given orally (5–10 mg every 12 h, with increas- dren, but the prevalence rises in older teenagers
ing dosage until high blood pressure is con- until the majority of patients present as adults.
trolled) for long-term therapy; i.v. or i.m. However, a child in a family with a diagnosis of
phentolamine (1 mg per dose) is used for hyper- this condition can be monitored and tested at a
tensive crises that may occur while the phenoxy- younger age. Manifestations have rarely been
benzamine block is being established. β-Blockade reported as young as 5 years of age. The syn-
by propranolol (5–10 mg given 3–4 times per day drome is due to loss-of-function mutations in the
orally for larger children) is added when the heart MENEN tumor-suppressor gene.
rate increases as the α-adrenergic blockade is Hyperparathyroidism will present in the usual
being established; propranolol may cause para- manner with elevated serum PTH and calcium,
doxical hypertension if given before the decreased serum phosphorus, and decreased bone
α-adrenergic block is established. If this therapy density. Localization may occur with a sestamibi
is ineffective, α-methyltyrosine (a tyrosine scan and treatment is accomplished by removal
hydroxylase inhibitor) can be started at 5–10 mg/ of the parathyroid glands.
kg/day, given four times per day. Nitroprusside is The gastrointestinal tumors of MEN type I
used as a vasodilator during surgical hyperten- are now termed gastroenteropancreatic-neuro-
sion. Surgical preparation also includes adminis- endocrine tumors or GEP-NETs subdivided into
tration of salt to repair the plasma volume, which carcinoid GEP-NET tumors that arise in the
is invariably low. lungs, GI tract, or thymus, while when they arise
260 10 Disorders of the Adrenal Gland

in the pancreas, the GEP-NETs are termed pan- 2B-associated mutations in the RET protoonco-
creatic NETs or pNETs. Gastrinomas in the duo- gene at 10q11.2) in contrast to the autosomal
denum lead to the Zollinger–Ellison syndrome dominant pattern of MEN 2A occurs usually with
of peptic ulcers. Insulinomas of the pancreas spontaneous mutations. Development of medul-
present and are treated in the manner noted in lary carcinoma of the thyroid is universal in the
Chap. 12. Glucagonomas raise blood sugar, so syndrome while about half of the affected patients
patient presents with what appears to be mild develop pheochromocytoma. MEN 2B is associ-
diabetes. The pituitary tumors occur at a younger ated with clear physical findings of skin and gen-
age than many of the other manifestations of eral body habitus. Ganglioneuromas of the lips
MEN1 and can secrete in decreasing order pro- and tongue are distinctive but can also occur on
lactin, growth hormone, and ACTH. The adrenal the conjunctiva, gastrointestinal tract, and urinary
cortical tumors of MEN1 are often nonfunc- tract. Failure to thrive in infancy occurs as well as
tional. Genetic testing of first-degree relatives megacolon, colonic diverticulosis, constipation,
of index cases of MEN1 is recommended after and diarrhea related to the GI ganglioneuroma.
genetic counseling. With a positive genetic diag- There is a marfanoid habitus with lax joints,
nosis, screening for the individual components is kyphosis, scoliosis, and lordosis as well as
performed regularly. hypotonia and developmental delay. Skin mani-
Multiple endocrine neoplasia type 4 (#610755 festations also include flushing. The physical
MULTIPLE ENDOCRINE NEOPLASIA, TYPE findings should cause the physician to obtain
IV; MEN4) presents a manner similar to MEN1 genetic testing and to search for occult pheochro-
and genetic testing should be carried out for this mocytomas and medullary carcinomas. The med-
variant if the patient has findings of MEN 1, but ullary carcinoma of the thyroid in this condition
genetic test for this condition is negative. often occurs at a younger age and is far more
MEN type 2A (#171400 MULTIPLE aggressive than found in MEN2A.
ENDOCRINE NEOPLASIA, TYPE II; MEN An experienced thyroid surgeon (a high-
2-associated mutations in the RET protoonco- volume surgeon usually carries out 30 thyroidec-
gene at 10q11.2) is an autosomal dominant con- tomies per year) must perform the thyroidectomy
dition which frequently has manifestations in the in children with various forms of MEN with MCT.
pediatric age group. Genetic testing for the muta- Von Hippel–Lindau disease (*193300 VON
tion in the RET gene is readily available and indi- HIPPEL-LINDAU SYNDROME; VHL at 3p26-
cated in all first-degree relatives of a proband. p25, 11q13) is an autosomal dominant condition
Because of the association with medullary carci- caused by a mutation in the von Hippel–Lindau
noma of the thyroid which often occurs earlier gene (VHL). Pheochromocytomas or gangliomas
and metastasizes at a younger age than is found may develop at a very young age. Adenocarcinoma
in sporadic medullary carcinoma of the thyroid, of the ampulla of Vater and pancreatic NETs and
children with positive genetic testing for MEN pancreatic carcinoma are also associated with the
2A should undergo prophylactic thyroidectomy. condition. Hemangioblastomatosis of the retina,
Pheochromocytoma is usually found in older cerebellum, and spinal cord are frequently found
patients, but it has been reported in children and in the syndrome. The condition is divided into
some recommend screening children with a posi- four subtypes of which three have pheochromo-
tive genetic test for MEN 2A starting at age 5. cytoma and one has renal carcinoma.
Parathyroid hyperplasia of MEN2 is almost uni-
versally found in adults with MEN 2A. The pheo- Neuroblastoma
chromocytomas are usually located in the adrenal Neuroblastomas are malignant tumors derived from
gland and may be bilateral or multicentric neural crest and are associated with excessive pro-
(see Chap. 14 for a discussion of the emergency duction of catecholamines; in spite of this, they rarely
management of pheochromocytoma). are seen with clinical symptoms of catecholamine
MEN type 2B (#162300 MULTIPLE excess (probably because of catecholamine catabo-
ENDOCRINE NEOPLASIA, TYPE IIB; MEN lism within the tumor), although a minority may have
Suggested Reading 261

hypertension. They often are diagnosed from their (Chrousos syndrome) and hypersensitivity. Endocr
Dev. 2013;24:67–85.
size or the presence of metastases and have a high
8. Stratakis CA. Cushing syndrome in pediatrics.
incidence of spontaneous regression. Urinary norepi- Endocrinol Metab Clin North Am. 2012;41(4):793–803.
nephrine (not epinephrine), VMA, and particularly 9. Simonetti GD, Mohaupt MG, Bianchetti MG.
HVA and dopamine are excreted in increased Monogenic forms of hypertension. Eur J Pediatr.
2012;171(10):1433–9.
amounts in neuroblastoma. Ganglioneuromas are
10. Auchus RJ. The physiology and biochemistry of
benign, mature forms of neuroblastomas derived adrenarche. Endocr Dev. 2011;20:20–7.
from sympathetic ganglion cells; usually silent with 11. Brown RJ, Kelly MH, Collins MT. Cushing syndrome
respect to endocrine function, they may manifest in the McCune-Albright syndrome. J Clin Endocrinol
Metab. 2010;95(4):1508–15.
some of the signs of pheochromocytoma.
12. Vehaskari VM. Heritable forms of hypertension.
Neuroblastoma is a finding in the Beckwith– Pediatr Nephrol. 2009;24(10):1929–37.
Wiedemann syndrome (#130650 ICD+ 13. Oskis A, Loveday C, Hucklebridge F, Thorn L,
BECKWITH-WIEDEMANN SYNDROME; Clow A. Diurnal patterns of salivary cortisol across
the adolescent period in healthy females.
BWS). It usually occurs in a sporadic manner due to
Psychoneuroendocrinology. 2009;34(3):307–16.
mutation in the imprinted gene at 11p15.5. 14. Waguespack SG, Ying AK. Chapter 14 -
Neurofibromatosis type I (#162200 ICD+ Pheochromocytoma and multiple endocrine neoplasia
NEUROFIBROMATOSIS, TYPE I; NF1) is caused syndromes. In: Sperling MA, editor. Pediatric endocri-
nology. 4th ed. Philadelphia, PA: Elsevier; 2014.
by mutation in the tumor–suppressor neurofibromin
p. 533–568.
gene (NF1) on chromosome 17. This condition is 15. Monticone S, Auchus RJ, Rainey WE. Adrenal disor-
associated with abnormalities of growth and puberty ders in pregnancy. Nat Rev Endocrinol. 2012;8(11):
(see Chaps. 5 and 9) and more rarely will be associ- 668–78.
16. Moisiadis VG, Matthews SG. Glucocorticoids and
ated with a pheochromocytoma. Physical findings
fetal programming part 2: mechanisms. Nat Rev
are café au lait spots and axillary freckling of the Endocrinol. 2014;10(7):403–11.
skin, Lisch nodules of the iris, and neurofibromas 17. Moisiadis VG, Matthews SG. Glucocorticoids and
and gliomas and the complications they cause. fetal programming part 1: outcomes. Nat Rev
Endocrinol. 2014;10(7):391–402.
18. Miller WL, Witchel SF. Prenatal treatment of congen-
ital adrenal hyperplasia: risks outweigh benefits. Am J
Obstet Gynecol. 2013;208(5):354–9.
Suggested Reading 19. Gamble KL, Berry R, Frank SJ, Young ME. Circadian
clock control of endocrine factors. Nat Rev
1. Brandon DD, Isabelle LM, Samuels MH, Kendall JW, Endocrinol. 2014;10(8):466–75.
Loriaux DL. Cortisol production rate measurement by 20. Nieman LK, Biller BM, Findling JW, Murad MH,
stable isotope dilution using gas chromatography- Newell-Price J, Savage MO, Tabarin A; Endocrine
negative ion chemical ionization mass spectrometry. Society. Treatment of Cushing’s Syndrome: An
Steroids. 1999;64(6):372–8. Endocrine Society Clinical Practice Guideline. J Clin
2. Martucci VL, Pacak K. Pheochromocytoma and para- Endocrinol Metab. 2015;100(8):2807–31.
ganglioma: diagnosis, genetics, management, and 21. Donohoue P. Diagnosis of adrenal insufficiency in
treatment. Curr Probl Cancer. 2014;38(1):7–41. children. http://www.uptodate.com/contents/diagnosis-
3. Miller WL, Flück CE. Chapter 13 - Adrenal cortex of-adrenal-insufficiency-in-children.
and its disorders. In: Sperling MA, editor. Pediatric 22 Donohoue P. Causes and clinical manifestations of
endocrinology. 4th ed. Philadelphia, PA: Elsevier; primary adrenal insufficiency in children. http://www.
2014. p. 471–532. uptodate.com/contents/causes-and-clinical-
4. Miller WL. Steroid hormone synthesis in mitochon- manifestations-of-primary-adrenal-insufficiency-in-
dria. Mol Cell Endocrinol. 2013;379(1-2):62–73. children.
5. Speiser PW, Azziz R, Baskin LS, Ghizzoni L, Hensle 23. Donohoue P. Treatment of adrenal insufficiency in
TW, Merke DP, Meyer-Bahlburg HF, Miller WL, children http://www.uptodate.com/contents/treatment-
Montori VM, Oberfield SE, Ritzen M, White PC. of-adrenal-insufficiency-in-children.
Congenital adrenal hyperplasia due to steroid 24. Donohoue P. Causes and clinical manifestations of
21-hydroxylase deficiency: an Endocrine Society central adrenal insufficiency in children http://www.
clinical practice guideline. J Clin Endocrinol Metab. uptodate.com/contents/causes-and-clinical-
2010;95(9):4133–60. manifestations-of-central-adrenal-insufficiency-in-
6. Santos L. Stress response in critical illness. Curr Probl children
Pediatr Adolesc Health Care. 2013;43(10):264–72. 25. Auron M, Raissouni N. Adrenal insufficiency. Pediatr
7. Charmandari E, Kino T, Chrousos GP. Primary gener- Rev. 2015;36(3):92–102.
alized familial and sporadic glucocorticoid resistance
Diabetes Mellitus
11

Diabetes mellitus (DM) is the second most Diabetes requires more of a patient or family
common chronic disease of childhood (if you do than most other chronic conditions and will often
not count obesity, asthma is the most common) exert significant psychosocial stressors to all
and is characterized by elevated blood glucose involved. This is a chronic condition in which
caused by inadequate insulin production or insu- everyday attention must be directed to (1) count-
lin action that leads to disordered carbohydrate, ing the carbohydrate composition of diet, (2) esti-
lipid, and protein metabolism. This may derive mating how much activity the child may engage
from inadequate insulin secretion (the basic in, (3) measuring and correcting the finger stick
defect in type 1 DM or T1DM), from inadequate blood sugar values at least four times a day, and
insulin action, or both (both defects occur in type (4) calculating and administering the correct insu-
2 DM or T2DM). Variations are found on these lin dosage usually at least four times a day. All this
two themes with some overlap in the presentation must be done without the child considering him-
of type 1 diabetes and type 2 diabetes. Until the self or herself “sick” as a child with diabetes really
last decades, type 1 DM was the most common should be considered as a healthy person who has
diagnosis in subjects younger than 18 years with to take care of his or her condition. The require-
new-onset DM, but now type 2 is becoming more ments of care may be difficult for some families to
prevalent and the incidence of T2DM exceeds the carry out as it requires some mathematical ability
incidence of type 1 DM in American Indian and understanding treatment concepts so they can
teenagers with the prevalence in other ethnic make medical decisions in the care of the child on
groups exceeding Caucasians rising. The differ- their own on a day-to-day basis. Divorce and sepa-
entiation between the two is not always straight- ration of parents is frequent in the present world;
forward in the young patient (Table 11.1). Recent communication between parents who are no lon-
statistics from four geographic areas of the USA ger together must remain seamless for the sake of
demonstrated a prevalence of type 1 diabetes in the child, but communication does not always
children of 1.93 per 1,000 with a rise of about continue in this manner. The pressures of the daily
20 % in the preceding decade. A more recent treatment regimen may lead to rebellion in teen-
study of commercially insured patients in the US age patients, and they may ignore the disease or
found a 57 % increase in the prevalence of type 1 may not be truthful in relating to parents or physi-
diabetes mellitus between 2002–2013 to 2.82 cians how they are treating themselves every day.
cases per 1,000. The prevalence of type 2 diabe- The treating physician has to be aware of all these
tes in children was 0.46 per 1,000 with a rise of issues and support the child and family as he or
30 % in the preceding decade. she addresses the complexities of this condition.

© Springer International Publishing Switzerland 2016 263


D.M. Styne, Pediatric Endocrinology, DOI 10.1007/978-3-319-18371-8_11
264

Table 11.1 Differential points between various common types of diabetes mellitus
Islet cell
antibodies,
insulin antibodies, Clinical characteristics or
Disorder Ketosis Insulin C-peptide GAD 65 antibody Inheritance Defect habitus
Type 1 Yes Low Low Positive Usually sporadic Autoimmune; Thin with weight loss but
diabetes beta cell failure during this obesity
mellitus epidemic may be heavy
Type 2 Sometimes Normal Normal Negative Familial Inherited insulin Overweight, possibly with
diabetes to high to high resistance and beta recent weight loss
mellitus cell failure worsened
with obesity
Atypical or Sometimes Normal Normal Negative Autosomal Mutation in the paired Occurs in African
ketosis prone to high to high dominant box gene 4 (PAX4) Americans
diabetes
mellitus or
Flatbush
Diabetes
MODY No Normal Normal Negative Autosomal At least 10 different Occurs before age 20 year
to high to high dominant genetic mutations Usually mild
hyperglycemia
MODY maturity-onset diabetes of the young
There are many more forms of Diabetes listed in the journal Diabetes Care and other sources
11
Diabetes Mellitus
Insulin Biosynthesis and Action 265

Insulin Biosynthesis and Action glucokinase is a rate-limiting step in the pro-


cess that leads to insulin secretion, it is
Insulin is synthesized by the beta cells of the islets described as the “sensor” for glucose in the beta
of Langerhans as proinsulin which is a single- cell. Glucokinase activity leads next to an
chain polypeptide. The C-peptide or connecting increase in the ratio of ATP/ADP which acti-
peptide is cleaved off of proinsulin leading to the vates the sulfonylurea receptor (SUR)-Kir6
two chains connected by two disulfide bridges of (inward-rectifying potassium channel) complex
the mature insulin molecule. The C-peptide is of the adenosine triphosphate-regulated potas-
produced only in the pancreas and is present in the sium channel KATP. Loss-of-function mutations
circulation only when endogenous insulin is of the KATP complex are responsible for hyper-
secreted. Thus it is a marker for endogenous insu- insulinemic hypoglycemia in infancy as
lin but not exogenous insulin. C-peptide may be described in Chap. 12 while gain-of-function
measured to indicate endogenous insulin secre- mutations lead to neonatal diabetes mellitus
tion in a state in which exogenous insulin is still and decreased insulin secretion described
present in the circulation; C-peptide will be absent below. An inactivating mutation in glucokinase
during surreptitious administration of insulin in is responsible for maturity-onset diabetes of the
Munchausen by proxy. young, or MODY type 3 (also known as mono-
Glucose enters the beta cell by action of the genetic diabetes type 3), which leads to a higher
glut 2 transporter and is then phosphorylated by threshold for insulin release and therefore rela-
glucokinase upon entry (see Fig. 11.1). Since tively mild hyperglycemia.

ATP-sensitive K+ channel
Ca2+
K+ Voltage
dependent
Ca2+ channel
SUR1
Depolarization
2+
Mitochondria Ca
6. R
KU
2

ATP/ADP↑ cAMP
Nucleus

Islet
Pyruvate transcription
factors
Glucose-6-phosphate
Glucokinase
Glucose
Secretory
granules Insulin

Glucose

GLUT2

Fig. 11.1 The beta cell and its components. The ATP- glucose-6-phosphate. Glucose-6-phosphate can be metab-
sensitive K+ channel (KATP) is the site of action of sulfo- olized during glycolysis leading to increased ATP which
nylureas which close the channel to cause insulin secretion enhances insulin secretion through the ATP-sensitive K+
and of diazoxide which opens the channel to suppress channel. Thus glucokinase gene acts as the glucose sensor
insulin secretion. Mutations in this area are responsible of the beta cell and plays a major role in determining how
for some forms of hyperinsulinemic hypoglycemia of the much insulin is released in response to glucose levels in
newborn. The GLUT 2 transporter allows glucose to enter the normal condition. Mutations in the glucokinase can
the beta cell where it interacts with glucokinase to produce lead to hypoglycemia or hyperglycemia
266 11 Diabetes Mellitus

Insulin secretion in response to elevated cose in the bloodstream. Insulin and glucagon are
glucose is biphasic. The first phase of insulin normally in balance to maintain normal blood
secretion after glucose stimulation is brief lasting sugar, but secretion is disordered in the hypergly-
for 2–5 min followed by a nadir at approximately cemia of diabetes. Glucagons, GLP 1 and GLP 2,
15 min. The second phase of insulin secretion are derived from preproglucagon encoded by the
occurs in about 2–3 h. Many studies aiming to preproglucagon gene which is expressed in the
predict the onset of diabetes implicate a decrease central nervous system, intestinal L-cells, and
in the first phase of insulin secretion as an initial pancreatic alpha cells. Posttranslational cleavage
defect indicating the development of diabetes. of preproglucagon leads to the production of
Insulin interacts with its membrane-bound mature glucagon in the alpha cells or GLP 1 and
receptor which is composed of four subunits and 2 in other cells depending upon which prohor-
is similar in structure to the IGF 1 receptor. A mone convertases (PC) are present. Glucagon
series of intracellular events follow when insulin receptors are seven-transmembrane G-protein-
interacts with its receptor, some of which are coupled receptors (see Fig. 1.4). When glucagon
affected by mutations which render the insulin couples with its receptor, activation of cAMP
receptor inactive. The intracellular processes occurs ultimately simulating the effects of gluca-
which occur after insulin or IGF 1 attach to their gon noted above.
respective receptors differ at physiologic levels. Most glucagon is produced and secreted by
However, there can be cross stimulation of either the alpha cells of the islets of Langerhans. GTP-
receptor at times of increased insulin or IGF 1 dependent potassium channels are triggered to
secretion. Insulin increases glucose uptake by release glucagon at low levels of glucose by acti-
adipose tissue and muscle directly through GLUT vation of the GTP-dependent potassium (KTP)
4 and into liver by more indirect manner as there channels similar to those found in the beta cell.
is no GLUT 4 on liver cell membranes. Insulin Glucagon secretion is also stimulated by amino
stimulates the production of glycogen in liver and acids, gastrin, and CCK in states of stress and
muscle and protein synthesis in muscle by the during starvation and exercise. Cortisol and
uptake of amino acids and stimulates lipid syn- growth hormone excess are associated with ele-
thesis through the uptake of triglycerides into vated serum glucagon. The secretion of glucagon
adipose tissue and liver. is inhibited by elevated blood sugar concentra-
In the fed state insulin rises due to oral glucose tions, GLP 1, free fatty acids, ketone bodies,
intake, while in the fasted state insulin secretion insulin, and, as stated above, somatostatin, an
decreases, but secretion of glucagon, growth hor- agent secreted from the delta cells with far-
mone, cortisol, and epinephrine, which are con- reaching effects including suppression of gluca-
sidered counterregulatory hormones that oppose gon release.
insulin action, rise. A discussion of the fed and Glucagon secretion is impaired in the early
fasted states is found in Chap. 12. stages of the development of diabetes leading to
a greater likelihood of hypoglycemia during
attempts to achieve “tight control.” Secretion of
Glucagon insulin from the beta cell may be a prerequisite
for local control of glucagon secretion. This may
While the action of insulin can be considered be a reason for the increased sensitivity to hypo-
anabolic in view of its effect of increasing glyco- glycemia as glucagon does not respond to low
gen stores in muscle and liver and lipids in adi- blood sugar when hypoglycemia occurs in the
pose tissue as well as enhancing the entrance of early stages of diabetes. However, glucagon is
nutrients into insulin-responsive tissues, gluca- elevated in other phases of the natural history of
gon can be considered a catabolic agent by diabetes. Glucagon is one of the factors that
enhancement of glycogenolysis. Glucagon stim- enhance the development of diabetic ketoacido-
ulates gluconeogenesis which also increases glu- sis (DKA) in the absence of insulin and promote
Other GI Hormones 267

the development of ketone bodies. In states of the medulla oblongata and GLP 1 also exerts
hypoglycemia in individuals with diabetes, glu- neurological effects by decreasing appetite, food
cagon is one of the factors that promote rebound intake, and water intake and increasing satiety.
hyperglycemia in the Somogyi effect, but in other GLP 1 is rapidly inactivated by cleavage at the
individuals with diabetes and hypoglycemic N-terminus by dipeptidyl peptidase-4 (DPP-4).
unresponsiveness, the glucagon response to GLP 1 agonists are used in the treatment of type
hypoglycemia which would ordinarily increase 2 diabetes in the adult to utilize the endocrine
glucose levels is sometimes defective. Since glu- effects of GLP 1 with the additional benefit of an
cagon promotes hyperglycemia, increased atten- extended period of activity. Likewise therapeutic
tion is now devoted to glucagon as a target for agents that inhibit DPP-4 and that serve as ago-
treatment of diabetes. nists for incretins are used to improve glucose
A glucagonoma is a rare finding in multiple control, but these are not approved for children.
endocrine neoplasia syndrome type I. GIP (gastric inhibitory peptide or glucose-
dependent insulinotropic polypeptide) stimu-
lates insulin secretion in response to glucose.
Other GI Hormones GIP is a 42-amino-acid polypeptide produced by
the K cells in the duodenum, proximal jejunum,
There are other significant influences on insulin and other areas of the small intestine.
secretion in addition to the effect of glucose. Carbohydrates and fats are the major nutrient
Insulin secretion is greater when glucose is stimulants for GIP secretion. GIP interacts with a
administered orally due to the effects of incretins, seven-transmembrane G-protein‐coupled recep-
hormones released from the gastrointestinal tract. tor similar to that for GLP 1 and glucagon.
It is estimated that incretin action causes 50–65 % Receptors are distributed in the pancreatic islets,
of the total insulin release that occurs in response stomach, brain, pituitary gland, lung, heart, and
to a meal. The incretin family includes glucose- vascular endothelium. Remarkably rare cases of
dependent insulinotropic polypeptide (GIP also food-dependent Cushing disease appear related
known as gastric inhibitory peptide) and to overexpression of GIP receptors in the adrenal
glucagon-like peptide (GLP noted above) which gland which are found in unilateral adrenal
stimulate glucose-dependent insulin release adenoma or bilateral macronodular adrenal
following oral ingestion. Glucagon-like peptide hyperplasia.
1 is produced in the intestinal L cells of the ileum Amlyn is a polypeptide secreted by the pan-
and colon by cleavage from the proglucagon creas along with insulin and has complementary
molecule by prohormone convertase PC1/3; thus effects on insulin. Amlyn slows gastric emptying,
the same gene codes for both glucagon and regulates postprandial glucagon, and reduces
glucagon-like peptide. GLP 1 is low in the fasting food intake in a similar manner to GLP 1 but does
state but rises after ingestion of food mainly stim- not stimulate the secretion of insulin. Pramlintide
ulated by glucose and fat. GLP 1 exerts its effect is an analog of amlyn which is available for the
by binding to a G-protein-coupled receptor on treatment of adults with type 1 diabetes, but treat-
the cell membrane of the pancreatic islets and ment with the native molecule produces amyloid
also has effects on the central and peripheral ner- fibrils which are harmful to the beta cell.
vous system, gastrointestinal tract, cardiovascu- Pramlintide is not approved for children.
lar system, kidney, and lung. The effects of GLP Ghrelin is an appetite-stimulating hormone
1 on the pancreas include enhancing secretion of (orexigenic) which is produced in several loca-
insulin and somatostatin and decreasing secretion tions in the body mainly on the gastrointestinal
of glucagon while enhancing beta cell prolifera- tract and also influences growth hormone secre-
tion. GLP 1 decreases gastric emptying time and tion via the growth hormone secretagogue
thereby brings about a sense of fullness. GLP 1 is receptor (GHS-R). Ghrelin is also found in the
also produced in the nucleus tractus solitarii of pancreas and appears to be involved in diabetes,
268 11 Diabetes Mellitus

but its role and action are controversial at pres- 60–70 % of acromegalic patients leading to the
ent. Ghrelin appears to suppress insulin secre- need for the development of new medications
tion and may be a diabetogenic factor. more specific to the type of receptor expressed on
Patients with severe lipodystrophy do not a given tumor. Since insulin secretion is sup-
produce leptin, the satiety hormone that origi- pressed by somatostatin produced by the delta
nates from adipose tissue. These patients have cells of the pancreatic islets, long-acting agonists
severe insulin resistance and long-term leptin of somatostatin are used in the treatment of some
treatment increases insulin sensitivity and thereby forms of neonatal hyperinsulinemic hypoglyce-
improves glucose levels and plasma triglycer- mia. Radiolabeled somatostatin analogs
ides. Leptin increases insulin sensitivity and (indium-111 (111In)-labeled pentetreotide) are
decreases insulin resistance. Leptin improves used to identify tumors with somatostatin recep-
blood sugar in type 1 and type 2 diabetes in ani- tors in adrenal medullary tumors (NEP tumors
mal models by suppressing the hypothalamic– including pheochromocytoma, neuroblastoma,
pituitary–adrenal axis, and this action may be ganglioneuroma, paraganglioma) or the GI tract
studied in the future in human beings as well. such as gastroenteropancreatic neuroendocrine
Somatostatin (also known as somatotropin tumors (carcinoid, gastrinoma, glucagonoma,
release-inhibiting factor (SRIF) or SS) is present vasoactive intestinal polypeptide-secreting tumor,
in a 14-amino acid peptide or a 28-amino acid pancreatic polypeptide-secreting tumor) and the
peptide depending upon cleavage of the pre- CNS (using technetium 99m depreotide [99mTc
prohormone. Somatostatin is produced in various depreotide], DTPA) for pituitary and extra sellar
sites including the delta islets of Langerhans of tumors. Excessive somatostatin secretion is found
the pancreas. Somatostatin exerts actions in somatostatinomas, rare tumors of the D cells of
throughout the body in addition to hypothalamic the islets of Langerhans which may be found in
somatostatin’s action of suppressing growth neurofibromatosis type I, multiple endocrine neo-
hormone secretion from the pituitary gland; thus plasia type I, or isolated occurrence. They may be
somatostatin suppresses the release of insulin, asymptomatic, but some of the patients manifest
glucagon, gastrin, cholecystokinin (CCK), vaso- the somatostatinoma syndrome which includes
active intestinal peptide (VIP), gastric inhibitory diabetes, gallstones, and steatorrhoea. The find-
polypeptide (GIP), secretin, motilin, and entero- ings of the syndrome might also be found with
glucagon. Somatostatin interacts with one of its treatment with somatostatin analogues.
five different G-protein-coupled receptors which Adiponectin appears to protect against adverse
cause differing effects. Thus, GH secretion is metabolic effects of obesity. Adiponectin
suppressed by SSTR2 and 5, insulin secretion increases sensitivity to insulin from a decrease in
is suppressed by SSTR5, and glucagon secretion hepatic gluconeogenesis and increases glucose
is suppressed by SSTR2. The SSTR2 receptor transport into muscles as well as increases energy
also affects the immune responses. Various consumption and fat oxidation in various sites.
tumors express differing somatostatin receptors; Tumor necrosis factor alpha (TNF alpha) is a
GH-secreting pituitary adenomas usually express pro-inflammatory cytokine that appears to play a
SSTR2 and SSTR5, prolactinomas express role in the development of both type 1 and type 2
SSTR1 and SSTR5, ACTH-secreting adenomas diabetes. Modulation of TNF alpha expression in
express SSTR5 with a lower abundance of animals can decrease the development of dia-
SSTR2 (low number), and gastroenteropancre- betes. Other inflammatory cytokines that appear
atic (GEP) neuroendocrine tumors usually to play a role in the development of type 1 and
express SSTR2 but others as well. The different type 2 diabetes are interleukin (IL)-1β, the IL-6
expression of receptors in various tumors is family of cytokines, and IL-18.
important for therapy; octapeptide somatostatin Pancreatic polypeptide (PP) secretion from
analogs, for example octreotide and lanreotide, pancreatic peptide-releasing cells (PP or F cells)
decrease growth hormone and IGF 1 in only of the islet of Langerhans increases during fasting,
Other GI Hormones 269

exercise, and hypoglycemia but is also stimulated tumors occur with multiple endocrine neoplasia
after protein, fat, and glucose ingestion. Pancreatic syndrome type I although more rarely than the
polypeptide secretion is decreased by somatosta- other classically described tumors.
tin, glucagon, and intravenous glucose. PP is Gastrin stimulates gastric acid secretion from
increased in pancreatic tumors also secreting insu- the parietal cells of the stomach and increases
lin and glucagon, gastrinomas, and Verner– gastric motility. Gastrin is released from G cells
Morrison syndrome (VIPoma); PP is used as a in the pyloric antrum of the stomach or more
marker for these tumors, now more commonly rarely in the duodenum and pancreas.
known as gastroenteropancreatic neuroendocrine Parasympathetic nervous stimulation leads to the
tumors (NET), when found with MEN1 or alone. release of gastrin-releasing peptide from the
Vasoactive intestinal polypeptide (VIP) is a vagus nerve which innervates the G cells.
28-amino acid peptide found mostly in neurons in Ingestion of amino acids into the stomach stimu-
the GI tract and the CNS among other sites. VIP lates the release of gastrin and bombesin also
interacts with its G-protein-coupled receptor on the stimulates gastrin release. While in the normal
cell membrane. VIP has many physiologic effects state gastrin is secreted from the stomach, tumors
as a neurotransmitter and neuromodulator includ- secreting gastrin are usually found in the pan-
ing affecting secretion and absorption in the GI creas or small intestine. Excessive gastrin pro-
tract, bicarbonate and fluid secretion from the bile duction is found in the neuroendocrine tumors
duct, relaxing smooth muscle of the esophagus, (NETs), gastrinoma which may occur in isolation
and inducing vasodilation. Because of its anti- or with multiple endocrine neoplasia type I
inflammatory anti-immunologic features, it may (MEN1): gastrinomas are the most common pan-
prove a therapeutic option for many conditions creatic tumors found in MEN1. Gastrinomas
including type 1 diabetes mellitus. A VIPoma cause the Zollinger–Ellison syndrome which
causes extremely high-volume diarrhea sometimes includes hypersecretion of gastric acid and ulcer-
known as pancreatic cholera, and measurement of ation of the duodenum or jejunum. Clinical find-
VIP levels will help in the differential diagnosis of ings in gastrinomas include abdominal pain and
this presentation. Radio imaging of VIP receptors heartburn. Weight loss occurs due to the GI
can be used to diagnose VIP-secreting tumors, pan- symptoms; associated diarrhea might suggest
creatic adenocarcinoma, and carcinoid tumors. VIPomas or carcinoid syndrome. Diagnosis is
Patients with Hirschsprung’s disease or achalasia accomplished by an elevated gastrin concentra-
appeared to have decreased VIP-containing cells tion during gastric acid secretion, by stimulation
and the GI tract. of gastrin secretion, or by functional studies of
Serotonin (5-hydroxytryptamine, or 5-HT) is gastric acid secretion.
mainly produced in the enterochromaffin cells of Chromogranin A is found in large dense core
the GI tract but also synthesized in serotonergic vesicles in neuroendocrine cells and is cosecreted
neurons of the CNS. In the GI tract it regulates with peptide hormones so that it serves as a
intestinal movement and in the central nervous marker for various neuroendocrine neoplasms.
system it regulates mood, appetite, and sleep. Thus elevated Chromogranin A is found in carci-
Serotonin also has a role in hemostasis and clot- noid tumors, islet cell tumors, pheochromocy-
ting when it is taken up by platelets. Serotonin is toma, pituitary tumors, and medullary thyroid
metabolized to 5-HIAA mostly by the liver. The carcinoma; elevated values are likewise found in
carcinoid syndrome is caused by an excess of hyperparathyroidism and hyperthyroidism.
serotonin and is characterized by flushing and Activation of beta 2 adrenergic receptors stim-
diarrhea and may be mistaken for a VIPoma ulates insulin secretion while blockade of these
although the diarrhea is less severe in carcinoid receptors decreases secretion. Activation of alpha
syndrome; the classic symptoms of carcinoid adrenergic receptors decreases insulin secretion
syndrome appear most closely related to meta- and blockade of alpha adrenergic receptors
static carcinoid tumors in the GI tract. Carcinoid increases secretion.
270 11 Diabetes Mellitus

Activation of the vagus nerve stimulates The prevalence of T1DM is 1.93/1,000 and of
insulin secretion. T2DM is 0.24/1,000 and the SEARCH for
Adiponectin is a cytokine which decreases in Diabetes in Youth study from the CDC and NIH
obesity and increases with healthy weight. reported in 2014 that over 191,000 children in the
USA have diabetes mellitus. The highest preva-
lence of type 1 diabetes is found in Finland with
Diabetes Mellitus the lowest in Venezuela. The prevalence in non-
Hispanic white youth is 1 in 370, in non-Hispanic
Absence of insulin or insulin action leads to dia- black youth is 1 in 438, and Asian/Pacific Islander
betes in all of its forms. Lack of insulin action is 1 in 1,250. Of newly diagnosed cases of diabe-
causes a catabolic state because of decreased tes mellitus, T2DM is found in non-Hispanic
entry of glucose into many cell types such as whites in 5.5 % of cases, in non-Hispanic black
muscle and adipose tissue through glut trans- in 37.6 %, in Hispanic in 35.2 %, in Asian/Pacific
porters which require insulin for their action (sg. Islander in 34.2 %, and American Indian youth in
GLUT4). There is subsequent impairment of 80 % of cases. There is an increase in the preva-
cellular action in these cells but not in the central lence of type 1 diabetes of approximately 3 % per
nervous system as the glut 1 transporter of the year in most countries including the USA. The
CNS requires no insulin to allow the entry of glu- incidence of diabetes mellitus is 28.1 per 100,000
cose. (See Chap. 12 for discussion of normal glu- per year. Every year over 18,000 children develop
cose metabolism.) In the absence of insulin effect T1DM while over 5,000 develop T2DM.
glucose rises since it cannot enter the cells
responsive to insulin. The renal threshold for glu-
cosuria in most people is 180 mg/dL, and as glu- Type 1 Diabetes Mellitus
cose rises above this level, glucosuria begins and
increases leading to an osmotic diuresis with loss Type 1 diabetes develops by destruction of the
of the calories ingested. This causes the classic beta cells of the islets of Langerhans (apoptosis)
findings of polyuria leading to dehydration with of the beta cells. Type 2 diabetes involves both
the individual development of polydipsia. apoptosis of the beta cells, initially to a lesser
Proteolysis and glycogenolysis proceed with degree, and insulin resistance. The pancreas
resulting weight loss. The absence of insulin in contains about one million islet clusters known
the portal circulation eliminates the inhibition of as the islets of Langerhans scattered throughout
the hepatic acyl carnitine cycle and thereby the pancreas and each of these islets comprises
allows the production of ketone bodies (acetoac- 1–2,000 beta cells. The islets are surrounded by
etate, acetone, and hydroxybutyrate). Elevated exocrine cells of the pancreas and ducts filled
ketone bodies lead to acidosis and cause shallow with proteolytic enzymes which can destroy the
rapid breaths in a hyperventilation condition islets after death or during pancreatic biopsy.
known as “Kussmaul breathing” which occurs in Islets are too small to image by available tech-
an attempt to eliminate carbon dioxide and niques, and thus postmortem studies are the main
oppose the metabolic acidosis. The presence of method of describing the degeneration of islets
ketone bodies will be noted on the breath as a cells during the progression of type 1 diabetes.
“fruity” smell, in the blood as ketonemia, and in Histologic studies reveal that CD8 T cells, cyto-
the urine as ketonuria. toxic T cells, play a major role in the develop-
Counterregulatory hormones (epinephrine, ment of insulinitis that occurs during the β-cell
glucagon, growth hormone, and cortisol) stimu- destruction found in type 1 diabetes. Macrophage
late gluconeogenesis, further increasing serum infiltration of the islets cells apparently also plays
glucose and worsening the problem. In the most a role. There is evidence that preproinsulin-
severe state of DM, DKA develops, with poten- specific CTL clones can kill β-cells. A prominent
tially fatal consequences, as described later. feature of type 1A diabetes is the presence of
Type 1 Diabetes Mellitus 271

autoantibodies to pancreatic beta cell antigens antigen (HLA) haplotype coded on chromosome
including glutamic acid decarboxylase 65 6 with a two- to threefold increased risk in indi-
(GAD65), insulin, IA-2 (islet cell-associated viduals with the HLA DR3 or DR4 haplotype
phosphorylase), and ZnT8 (zinc transporter and a five- to sevenfold increase in risk with both
molecules). Detection of these antibodies assists haplotypes in the same person. Type 1 DM is
in the classification of the type of diabetes an 100 times more frequent in those with a missing
individual may have and assists in the prediction aspartic acid at position 57 in the DQ β chain
of the development of type 1 diabetes in clinical than when there is an arginine at position 52 of
studies in a first-degree relation to a proband with the DQ α chain. The risk of onset of type 1 dia-
diabetes who is presently euglycemic. However, betes mellitus in nonidentical siblings is 1 % if
type 1B diabetes (or idiopathic DM) has no circu- they share no HLA D haplotype, 5–7 % if they
lating antibodies measurable. The report of a share one haplotype, and 12–20 % if they share
patient with X-linked agammaglobulinemia who both haplotypes. Other HLA allele haplotypes
can make no antibodies but nonetheless devel- reduce the likelihood of the onset of type 1 DM;
oped type 1B diabetes emphasizes that antibody for example an aspartic acid at position 57 in a
formation is not a necessary step in the develop- heterozygous pattern (non-Asp/Asp) decreases
ment of diabetes mellitus. In contrast to beta cell the risk. For Caucasian children, a 6 % risk exists
destruction which is characteristic of diabetes, for a sibling of an affected proband to develop
alpha cells which produce and secrete glucagon type 1 DM if the proband is younger than 10
remain as do delta cells which produce and years, and a 3 % risk if the proband is older than
secrete somatostatin. 10 years. Further, a 2–5 % risk of type 1 DM is
Type 1 diabetes mellitus (*222100 DIABETES found in an offspring of a diabetic parent, the
MELLITUS, INSULIN-DEPENDENT; IDDM), higher risk occurring if the father is affected. A
previously called juvenile-onset diabetes melli- full explanation of the genetic basis for type 1
tus, is a state of insulin deficiency requiring insu- diabetes eludes us but it is clear that a genetic
lin replacement for therapy; this condition cannot predisposition modified by an environmental
be treated with oral hypoglycemic agents. This exposure is the etiology.
classification is divided into type 1A diabetes of Many environmental factors are suspected to
autoimmune basis which occurs in an individual play a role in the onset of type 1 DM. The onset
with a genetic susceptibility who encounters an of type 1 DM is more frequent in Fall and Winter
environmental stress and represents the majority in either hemisphere (suggesting a link to viral
of cases. Type 2B diabetes has similar findings, infections, which are more prevalent then) but
but no laboratory tests support an autoimmune can occur all year. The link to viral infections is
basis. In this chapter we will refer to type 1 diabe- suggestive, but the only definitive link of type 1
tes mellitus but will usually be directing com- diabetes with viral infection is with congenital
ments to type 1A diabetes. rubella. Besides viral infections, other factors
Type 1 DM does not only occur in childhood such as environmental chemical substances are
as half of the cases are diagnosed in the adult. considered as a trigger for type 1 diabetes in sus-
There is a 50 % concordance rate of type 1 dia- ceptible individuals. However, there is no con-
betes mellitus in monozygotic twins, indicating clusive information at present of what exposure
that environmental effects are of importance in is important, and it is impossible to avoid expo-
addition to genetic factors or the concordance sures that are linked to the development of type 1
would be 100 %; 100 % is the approximate con- DM. Several ongoing studies on cohorts starting
cordance rate of type 2 diabetes mellitus in at birth aim to determine environmental factors
monozygotic twins demonstrating the stronger that might be involved in the development of
genetic tendency of this condition. Relationships T1DM. Other research studies first-degree rela-
exist between the likelihood of developing type tions of probands with T1DM to determine
1 diabetes mellitus and the human leukocyte potential etiologies and determine if it is possible
272 11 Diabetes Mellitus

to delay or eliminate the development of T1DM After the onset of clinical type 1 DM and the
in those individuals. start of therapy, there is often a “honeymoon
As noted, type 1-A diabetes is the specific period” of recovered beta cell function and
diagnosis given to the autoimmune form of type decreased insulin requirements, which may,
1 diabetes which is the form most often encoun- incorrectly, suggest cure of the disease process
tered. About 90 % of individuals with type 1 DM and may last months to a year or more. Clinical
have positive diabetes-related antibodies con- trials of immunosuppressive therapy to lengthen
comitant with the discovery of clinical hypergly- the honeymoon period and other studies have
cemia. Glutamic acid decarboxylase 65 (GAD been undertaken to determine how to avoid the
65) converts glutamate to GABA which is found development of type 1 DM.
in the nerves terminating at the beta cell and anti-
bodies to GAD 65 are commonly found in type
1A diabetes mellitus as are antibodies to insulin, Clinical Presentation of Diabetes
islet cells, IA-2 (islet cell-associated phosphor- Mellitus
ylase), and ZnT8 (zinc transporter molecules).
Since an increase in the titer of these antibodies Polyuria, polydipsia, and weight loss with dehy-
precedes the onset of clinical type 1 DM by dration are the classic findings of all types of DM
months to years, it has become clear that a and may be noted, usually in retrospect, for sev-
sequence of beta cell destruction ultimately leads eral weeks before presentation. If the family or
to the clinical picture that we recognize as type 1 physicians are attuned to the findings, the diagno-
DM. Months pass after exposure to the initial sis might be made before the development of the
factors that start the process in a susceptible DKA that is the inevitable consequence of severe
patient until the beta cell function is reduced by untreated insulin deficiency; diabetes is so com-
80 % to about 20 % of normal levels, which is the mon in the present era that a relative will often
degree of impairment that leads to clinical mani- measure the blood sugar of an ill child in the
festations of diabetes. An intercurrent infection home, leading to the discovery of hyperglycemia.
or other acute stress may precipitate the onset of Thus serum glucose might be moderately ele-
type 1 DM or DKA after a period of subclinical vated at presentation (e.g., 200–300 mg/dL)
impairment of beta cell activity is determined. without the presence of substantial serum ketones
The autoimmune basis of type 1 DM also or decreased mentation, or conversely, the serum
predisposes the affected individual to other glucose might be severely elevated at presenta-
autoimmune diseases including Hashimoto thyroid- tion (700 mg/dL or more) with severe ketoacido-
itis, autoimmune Addison’s disease, autoimmune sis (pH decreased to 6.9 or lower) and stupor or
ovarian disease, and celiac disease. Type 1 DM coma, all depending on when the natural course
may be included in autoimmune polyendocri- of the disorder is interrupted with medical evalu-
nopathy syndrome 2 (269200 SCHMIDT ation and care. About 25 % of subjects are seen in
SYNDROME), which may include candidiasis clinically apparent DKA at the onset of the con-
along with impairment of endocrine glands (see dition, as described later.
Chap. 10). Autoimmune polyendocrinopathy In the presentation of T1DM in this obesity
syndrome #3 may include type 1 diabetes along epidemic, the classic thin appearance of
with Hashimoto thyroiditis. new-onset T1DM may not occur. In some cases
The increase in such antibodies is utilized in unexplained weight loss is viewed as a positive
clinical research studies of first-degree relatives sign in a child with obesity when in fact it is an
of a proband to predict the onset of clinical DM, early manifestation of developing diabetes melli-
especially if the antibody titer is combined with a tus. The abdominal pain and nausea and vomiting
measure of impaired beta cell function. If three characteristic of DKA might instead be mistaken
antibodies are present in the relative, there is a for a gastrointestinal disorder. The rapid
high likelihood of the development of diabetes Kussmaul breathing might be mistaken for respi-
within 5 years. ratory disease. The obtundation that may develop
Type 1 Diabetes Mellitus 273

may be mistaken for substance abuse. Glycosuria symptoms noted earlier, with repetition of these
might be mistaken as a sign of a reduced renal laboratory findings on at least a second occasion.
threshold for glycosuria (because some otherwise If frank DKA is found, the diagnosis requires no
normal individuals do have a renal threshold as further blood sugar confirmation. It must be noted
low as 150 mg/dL) or as the presence of renal that even a fasting blood sugar (BS) of 126 mg/dL
tubular disease. Diabetes is a relatively common is outside of the normal range which extends only
disorder and must always be kept in mind in the to 100 mg/dL; a fasting blood sugar of 100–125 is
evaluation of the child showing these various considered “impaired fasting glucose” and a 2-h
symptoms and signs. An elevated blood sugar postprandial value of 140–200 is considered
sometimes occurs with glycosuria in the presence “impaired glucose tolerance” and either or both
of severe infection or trauma, but the findings indicate the development of prediabetes. The
remit along with the primary illness. In some ADA has recently established a hemoglobin A1c
cases these children will indeed develop bona ≥6.5 % as a diagnostic criteria for diabetes and
fide diabetes later and should be watched espe- prediabetes (5.7–6.4 %). However, hemoglobin
cially if there is a family history of diabetes; type A1c values tend to be variable in teenagers in chil-
2 diabetes is more commonly found in multiple dren and many caring for children use the hemo-
members of the family than type 1 diabetes as is globin A1c as supportive information rather than a
monogenetic diabetes (maturity-onset diabetes of tool for the definitive diagnosis (Table 11.3).
young, or MODY). The finding of the “shift to An elevated titer of antibodies to islet cells,
left” in the blood count which is characteristic of insulin, or GAD 65 is found in the serum in 90 %
DKA may be interpreted as an isolated infection. of cases of type 1 diabetes. Urine analysis will be
Since many children with bona fide new-onset
diabetes mellitus will present with infections that
hastened the onset of the clinical presentation, the Table 11.2 Glucose tolerance test after 1.75 g/kg carbo-
hydrate load (75 g maximum)
clinician must be careful to differentiate between
the two situations. Condition Baseline 1h 2h 3h
If diabetes is considered and glycosuria and/or Normala <110 <200 <140 <130
an elevated finger stick blood sugar is determined Insulin 9 51 37 20
(mU/mL)
by glucometer, it is never appropriate to send a
C-peptide 1.3 3.3 3.0 2.0
confirmatory laboratory glucose determination (ng/mL)
which cannot return on the same day; the author Impaired 110–125 140–199
has seen cases in which this sequence has occurred glucose
over a 3-day weekend so that when the glucose tolerancea
value, well above 200 mg/dL, returned days later Diabetes ≥126 ≥200 ≥200 ≥200
mellitusa
from the laboratory, the child was already in the
a
intensive care unit being treated for DKA. If dia- Blood glucose in mg/dL
betes is considered as the diagnosis, further diag-
nostic activities must occur immediately. Table 11.3 HgbA1c compared with average blood sugar
Laboratory evaluation will reveal elevated HgbA1c (%) Average blood sugar (mg/dL)
serum glucose in diabetes mellitus in the presence 4 60
of polyuria and polydipsia and a glucose tolerance 5 90
test (GTT) is not required in this situation 6 120
(Table 11.2 has standards for GTT for use in other 7 150
circumstances for completeness but is rarely uti- 8 180
lized). The diagnosis, according to the American 9 210
Diabetes Association, specifically requires a fast- 10 240
ing blood glucose equal to or greater than 126 mg/ 11 270
dL or a random (or casual) blood glucose greater 12 300
than 200 mg/dL, with the presence of the 13 330
274 11 Diabetes Mellitus

positive for glucose, as the elevation of blood ketonuria with urinary ketones present, usually
glucose will exceed the renal threshold of “large,” along with acidosis defined as venous
180 mg/dL that allows glucose excretion into the blood gas pH less than or equal to 7.30 and bicar-
urine; however, glycosuria also can occur in bonate equal to 15 mEq/L or less. The level of aci-
nondiabetic conditions such as a genetic decrease dosis defines the severity of diabetes so that mild
of the renal threshold for glucose, diseases that DKA is characterized by venous pH of 7.2–7.3 or
affect the renal tubule, times of severe illness and HCO3 10–15 mEq/L, moderate DKA as pH 7.1–
stress, and even on occasion after excessive 7.2 or HCO3 5–10 mEq/L, and severe DKA as pH
ingestion of sugar. Positive urine or blood ketone <7.1 or HCO3 less than or equal to 5 mEq/L
bodies (acetoacetate, acetone, hydroxybutyrate) (Table 11.4). Besides the usual symptoms of poly-
will develop in the absence of insulin, but ketone dipsia and polyuria, weight loss may accelerate
bodies also may be found at presentation in chil- because of fluid losses caused by osmotic diuresis
dren with type 2 DM, making differentiation and vomiting. Abdominal pain is common, some-
between the two types difficult. Because of the times suggesting the incorrect diagnosis of acute
elevated serum glucose, the serum sodium will abdomen. Patients at presentation usually have
be low, because of loss of sodium due to osmotic severe elevation of blood glucose, metabolic aci-
diuresis and because a 1.6-mEq/L decrease in dosis with ketone bodies in blood and urine, and
serum sodium concentration is found for every dehydration with urinary sodium and potassium
increase in serum glucose of 100 mg/dL. Insulin loss. Dehydration causes increased serum creati-
deficiency leads to elevation of serum triglycer- nine and blood urea nitrogen (BUN; factitious
ide and cholesterol, sometimes to a remarkable elevation of creatinine may also occur because of
degree of over 1,000 mg/dL and several 100 mg/ cross-reaction of acetoacetate in the creatinine
dL, respectively. These lipid abnormalities may assay) and raises blood sugar even more.
precipitate pancreatitis. The percentage of hemo- Dehydration increases acidosis. The respiration
globin A1c (HgbA1c) demonstrates the degree to rate is increased to counteract the metabolic acido-
which glucose is nonreversibly bound to Hgb and sis, and the ketonemia adds a “fruity” aroma to the
provides a window into the length of time the breath; deep-“sighing” Kussmaul respiration is the
child has been hyperglycemic. term used to describe the rapid but shallow respi-
rations. A patient with type 1 DM might certainly
Diabetic Ketoacidosis have intercurrent pneumonia, but Kussmaul respi-
DKA may be the presenting condition at diagno- ration does not by itself indicate pneumonia; con-
sis of diabetes. Risk factors for the development versely, the state of dehydration may minimize the
of DKA are age under 5 years, lower socioeco- signs of pneumonia, so consideration for this pos-
nomic and educational status, and lack of access sible coexisting condition must occur during eval-
to medical care. But DKA is not limited to the uation of DKA. Indeed, infection may be present
first episode in a previously undiagnosed patient, and may have been the precipitating factor to
as DKA may develop in any patient who does not cause worsening of the diabetic condition, so the
adequately care for the condition or misses sev- possibility of infection must be considered in all
eral insulin injections. cases of DKA. Even if no infection is present, the
While DKA is known to be related to T1DM, white cell count is often elevated in DKA, with a
DKA may likewise occur in type 2 diabetes in marked shift to the left and prevalence of band
children in contrast to the absence of DKA in most forms of white blood cells (WBCs), related to the
adults with type 2 diabetes. DKA may be defined elevation of glucocorticoids during stress.
as a condition where the glucose is greater than Metabolic acidosis may be demonstrated if the
200 mg/dL, there is ketonemia determined by a anion gap (calculated as [Na+–(HCO3− + Cl−)]) is
serum beta-OH butyrate greater than 4 mEq/L or increased above the normal limit of
ketones strongly positive by Ketostix measure- 14 mEq/L. Tachycardia is common. The patient
ment at a greater than 1:2 dilution of serum, may be confused, stuporous, or in frank comma.
Type 1 Diabetes Mellitus 275

Table 11.4 The treatment of DKA as carried out at UC must be modified along with medication doses as needed
Davis Medical Center developed by the pediatric endo- by the clinical situation encountered
crine, PICU and ED teams; this is only a guideline that
Diabetic treatment plan, pediatrics
Standard admission orders for PICU
1. Admit to PICU
2. Initiate diabetes education
3. Diet: NPO
4. Activity as tolerated
5. I & O and daily weight
6. Lab work:
ABG or VBG gas with electrolytes on admission and then q 3 h
BMP with calcium, phosphate, magnesium on admission and q. 6 h
Amylase and lipase on admission
Blood glucose monitoring q 1 h until i.v. insulin is discontinued
Check urine ketones every void once subcutaneous insulin started
7. Administer O2 to keep O2 saturation >95 % if necessary
8. IV fluids:
Calculations:
Deficit: Assume 7 % dehydrated; weight (kg) × 70 = (a) _____mL
Remaining deficit; subtract all boluses given: (a) − bolus IVF = (b) _____mL
Replace remaining deficit over 48 h: (b)/48 h = (c) _____mL/h
Maintenance IVF: (d) _____mL/h
Total IVF (maintenance + remaining deficit): (c) + (d) = _______mL/h
Serum glucose >250 mg/dL
Normal saline with 20 mEq KCl and 20 mEq KPO4 at total IVF rate
Serum glucose <250 mg/dL
D10 ½ normal saline with 20 mEq KCl and 20 mEq KPO4 at ½ total IVF rate plus ½ normal saline with 20 mEq
KCl and 20 mEq KPO4 at ½ total IVF rate to make D5 ½ NS with 20 mEq KCl and 20 mEq KPO4 at total IVF rate
(Note: there is controversy over whether normal saline should be used in almost all situations including this. You
must use clinical judgment in the condition of the patient to decide on this issue)
9. If urine output not >1 mL/kg/h or serum K >5.0, call MD immediately
10. Regular human insulin continuous infusion at 0.1 U/kg/h
11. Once insulin infusion is begun, call MD if serum glucose is <150 mg/dL or >250 mg/dL
12. Acetaminophen 15 mg/kg = _________ q 4 h PO/PR PRN temperature >38.0 or headache
13. VS, neuro checks, and cardiac monitoring q 1 h while on i.v. insulin, then per unit routine. If GCS < 13 or if
patient c/o headache, neuro checks q ½ h and call MD
14. If it is anticipated that the patient will be transitioned to SC insulin in a.m., please order an UNLIMITED ADA
breakfast
15. When patient is transitioned to SC insulin, home glucose monitor may be used for blood glucose testing
Note: If the patient has known diabetes and is on an insulin pump, the pump should be disconnected and the infusion
set removed. The parents should be instructed to bring a new infusion set to the hospital the following day so that the
pump can be restarted after the patient recovers from DKA

DKA may be the first presentation of diabetes were performed. DKA presenting after the diag-
in an individual. Sometimes the child has visited nosis of diabetes however is frequently due to
doctor within the previous week or two and the missed insulin injections, sometimes purpose-
more subtle symptoms were not identified as fully. Teenage girls may note that they are gain-
indicative of diabetes and no diagnostic tests ing weight on insulin therapy and wish to lose
276 11 Diabetes Mellitus

weight by omitting the insulin. Teenagers may respiratory arrest, are the most extreme presenta-
act out by skipping their insulin injections or not tions. A patient who appears to be improving but
attending to their insulin pump appropriately. deteriorates, or who develops a headache during
DKA may occur with unrecognized insulin pump the improvement phase, or younger children who
infusion malfunction, usually due to the infusion develop uncharacteristic behavior such as crying,
set being kinked or clogged or having fallen out blinking, or restlessness may be developing clini-
of the skin without being noticed. cally relevant CE. Early signs of increased intra-
However, DKA may also occur because of cranial pressure include (a) altered mental status/
severe intercurrent illness. Most families edu- headache with decreased papillary response to
cated in diabetes management can treat mild to light, (b) bradycardia, (c) hypertension, and (d)
moderate ketonemia at home with appropriate irregular respiration, usually developing later in
fluid and insulin management as long as the the course. Treatment must be rapid and may
child can drink and eat, but if the child has severe include hyperventilation, mannitol infusion
diarrhea or severe vomiting, a visit to the emer- (0.25–1 mg/kg i.v., given over 20 min every
gency department or admission to the inpatient 2–4 h), and possibly glucocorticoid administra-
ward is likely to be necessary. All children with tion, although the glucocorticoid may itself
diabetes should receive a yearly influenza vac- increase blood glucose values. Hypertonic 3 %
cine injection (not nasal spray which is not saline has been used less frequently in a dose of
approved for diabetes); influenza infection will 5–10 mL/kg over 30 min, but there are few stud-
very likely lead to hospitalization and need for ies of this approach. Elevation of the head may
intravenous fluids. Present recommendations help. Central nervous system (CNS) imaging is
suggest that every child with diabetes who is the direct proof of the condition and should be
over 2 years old should get pneumococcal immu- performed if imaging can be carried out while the
nization as well. subject is carefully observed and emergency ther-
The life-threatening complication, cerebral apy is available. Neurosurgical consult is manda-
edema (CE), occurs in about 1 in 100 cases of tory, as cranial decompression may be necessary.
DKA, more often in younger patients, and may The cause of CE is in dispute, and the classic
occur during the hospitalization at which the ini- view that an excessive rate of rehydration is the
tial diagnosis of type 1 DM is made. Mortality is primary cause leading to cerebral edema is not
reported in 20–40 % of cases, and neurologic supported by recent data. However, careful atten-
sequelae in survivors are reported to be about tion to the rate and composition of fluid replace-
25 %. A recent study of 6,000 cases of DKA indi- ment in diabetic ketoacidosis by following
cated that CE is more common in patients having established guidelines is a prudent measure.
the lowest serum bicarbonate levels, in patients Other possible complications of DKA include
who received bicarbonate administration, in the development of thrombosis or pulmonary
those with the youngest ages, in those with the embolism and renal insufficiency due to throm-
longest duration of symptoms and in those with bosis of major vessels. Thus although the treating
the highest severity of symptoms, in those with physician must endeavor to moderate fluid
higher BUN, and in those without the normal administration so as not to increase the likelihood
increase in serum Na during therapy. CE may of CE, fluid must be adequate to avoid these com-
present with a depressed sensorium from 12 up to plications of severe dehydration.
24 h after presentation of DKA and after initial Total body potassium loss may not be directly
improvement of symptoms. However, many reflected in serum levels of potassium as potas-
patients with any degree of DKA have some sium is an intracellular ion. Thus hyperkalemia
degree of brain swelling (which can be demon- or hypokalemia may occur during DKA or dur-
strated on magnetic resonance imaging (MRI)) as ing its treatment. EKG monitoring therefore is
a manifestation of subclinical cerebral edema, important during evaluation and treatment of
but the clinical manifestations of CE, including DKA as T waves are peaked in hyperkalemia and
cerebellar tonsillar herniation and subsequent low in hypokalemia with U waves appearing.
Type 1 Diabetes Mellitus 277

Monitoring and Treatment of Diabetic of headache, bradycardia, change in neurological


Ketoacidosis (Table 11.4) status including such findings as restlessness,
DKA is a life-threatening emergency that requires irritability, increased drowsiness, incontinence,
close monitoring to be carried out in a pediatric and the development of specific neurological
intensive care unit, whenever possible. Patients signs such as cranial nerve palsies, rising blood
from smaller communities are often transported pressure, and decreased oxygen saturation.
to such centers by air or ambulance for this care. Because of the potential development of cere-
Alternatively pediatric intensivist or pediatric bral edema, hypertonic (3 %) saline and mannitol
emergency department physicians can provide must be kept at hand and used should cerebral
guidance to rural providers via telemedicine from edema develop.
major medical centers. A recent consensus con- Vital signs and mental status are checked
ference of the international Society for Pediatric hourly in affected patients. Blood glucose is mea-
and Adolescent Diabetes developed evidence- sured hourly; a true laboratory value is best, but a
based guidelines for DKA which are summarized bedside glucometer is frequently used.
below: Glucometers must be tested for accuracy, and the
Standard evaluation by the pediatric advanced age of the sensor strips used on the device must
life support guidelines is carried out with deter- be checked for expiration dates to make sure that
mination of blood gas, ketones, CBC, metabolic they are not too old, which would make them
panel and state of dehydration, state of conscious- more likely to report incorrect values. As BS val-
ness, and the establishment of two i.v. lines. ues descend to the normal range, the glucometer
Fluid replacement should be the first treat- becomes less accurate, and laboratory confirma-
ment but complete replacement should occur tion of glucose values becomes even more impor-
slowly over 36–48 h. tant. Serum electrolytes are measured every 4 h.
Monitoring for the development of cerebral Serum osmolality is measured in the laboratory,
edema includes observation for the development but in the interim may be roughly estimated as

mOsm / L = 2 ´ serum sodium ( mEq / L ) + éë blood glucose ( mg / dL ) / 18 ùû .

This equation indicates that an increase in The amount of fluid loss in DKA is estimated
blood glucose of 180 mg/dL causes an increase in according to customary clinical findings. Severe
osmolality of 10 mOsm/L. Thus DKA is a condi- cases are usually associated with fluid loss equiv-
tion of hypertonic dehydration, and osmolality is alent to 10–15 % of body weight, but the standard
elevated at presentation. presentation is more likely 7 % dehydration.
Treatment of DKA involves fluid and electro- Clinical signs of dehydration are as follows: 5 %,
lyte resuscitation along with insulin administra- normal skin turgor, moist buccal mucosa, tears
tion. Careful attention to laboratory data and fluid present, and pulse regular; 10 %, skin dry, and
administration is accomplished with a flow sheet tenting of skin possible, buccal mucosa, dry eyes,
kept up to date at all times; such a flow chart is deep set, tears decreased, irritability noted, and
not an option. The concern that excess fluid pulse increased; and 15 %, clammy skin, parched
administration may predispose to complications buccal mucosa, eyes sunken, no tears, lethargy
must be balanced by the possibility that the noted, and pulse rapid. Because of the osmotic
dehydration at the time of presentation may cause diuresis, urine output is not a reliable indicator of
ischemia of kidney, brain, or other organs. Thus hydration.
any guideline for therapy must be modified by Most recommend that normal saline solu-
the clinical condition of the patient. tions be used in the initial phases of rehydration,
278 11 Diabetes Mellitus

and there is increasing evidence that hypotonic vital signs [low blood pressure (BP), rapid
fluid leads to complications in many types of pulse, and delayed capillary filling], and this
dehydration including DKA. Fluid should be bolus may be repeated if indicators suggest the
replaced slowly, over a 36–48-h period, accord- need for more immediate fluid replacement. If
ing to most recommendations, with half given the patient is in shock, blood or plasma expand-
over the first 12 h, and the rest, over the remain- ers may be used according to customary inten-
ing 24–36 h. Initial resuscitation may be offered sive care protocols.
as a bolus of normal saline at 5–10 mL/kg if sig- Sodium deficits develop with DKA and are
nificant dehydration is detected by abnormal calculated as

mEq of sodium required = éëconcentration desired ( mEq / L ) - concentration present ( mEq / L ) ùû


´ 0.6 - 0.7 ´ body weight in kilograms

One half of this deficit is given in the first 12 h Acidosis is almost always corrected by fluid
and the rest in the next 24–36 h. An additional and insulin administration in DKA. Thus added
20–40 mEq/L of sodium is added for daily bicarbonate is not indicated in most cases, even
maintenance. with significant acidosis; indeed, added bicar-
Potassium deficit is present in all presenta- bonate is associated with an increased likelihood
tions of DKA. While potassium is mainly an of the onset of cerebral edema and may paradoxi-
intracellular ion, a serum potassium value that is cally cause cerebral acidosis. While some author-
normal indicates a moderate deficit in total body ities recommend the use of 40 mEq/m2 of
potassium, whereas a low value indicates a serve bicarbonate if the pH is 7.1 and 80 mEq/m2 if the
deficit, and even an elevated value is associated pH is less than 7.0, this chapter does not advo-
with a mild potassium deficit. Potassium deficit cate routine bicarbonate administration. For
will increase with saline administration and completeness only we include the formula to cal-
expansion of extracellular volume. As acidosis is culate bicarbonate deficit as (12 − observed bicar-
corrected, or if alkalosis develops, the potassium bonate value) × 0.6 body weight (kg), and the
will leave the intracellular space in exchange for administration is performed as 1–2 mEq/kg over
hydrogen, a trend that intensifies with the a 2-h period or slower.
glucose and insulin administration in the course Insulin therapy for DKA is generally adminis-
of the treatment. Careful attention to potassium tered as an i.v. infusion at a rate of 0.05–0.1 U/
balance is essential. Although laboratory evalua- kg/h, with the lower dose range usually achieving
tion is essential, the electrocardiogram (EKG) a more desirable slower decrease in blood glu-
offers some information, as peaked T waves are cose. We do not advocate administration of a
seen in hyperkalemia, whereas U waves are seen bolus of i.v. insulin initially to saturate sites on
in hypokalemia along with low T waves. the infusion set, as was the course in the past. The
Potassium can be replaced, once urinary output is goal of insulin therapy is a decrease of blood glu-
established, by 20–40 mEq/L given slowly in the cose of about 50–100 mg/dL/h. Correction of
replacement fluid. EKG monitoring is essential, blood glucose may precede correction of acid–
as an excessive amount or speed of potassium base balance. In this situation, it is important to
administration may be fatal because of the devel- continue insulin administration until acidosis is
opment of arrhythmias. Because Cl may itself eliminated. Although no dextrose is given in i.v.
cause acidosis, potassium is best given half as fluids at first, as the blood glucose is already
potassium acetate and half as potassium phos- elevated, when blood glucose decreases to about
phate, with the proviso that phosphate infusion is 300–200 mg/dL, dextrose (usually as D5–10 %
best used only when phosphate is low, because in in saline) is added to allow the safe, continued
excess, phosphate can reduce calcium. administration of insulin, which will serve to
Type 1 Diabetes Mellitus 279

eliminate ketone bodies while minimizing the and if glucose is only moderately elevated. It is
likelihood of hypoglycemia. important but sometimes difficult to differentiate
Intravenous insulin is now always administered this situation from type 2 DM described later.
in DKA as recombinant DNA-derived regular Parents will not be in the best emotional condi-
human insulin. IV insulin is administered via a tion to learn of the care of the child with diabetes
“Y” connector piggybacked onto the other fluids and outpatient management may be efficient but
being administered. Insulin is usually mixed as inappropriate for some situations.
50 U into a 250-mL bottle of saline, yielding 1 U
insulin/5mL of saline. The desired hourly units of Transitioning to the Long-Term
insulin will be delivered by the number of millili- Management of Diabetes Mellitus
ters determined by multiplying the units by 5 (e.g., Type 1
2 U/h is given as 10 mL/h insulin solution). The Once the diagnosis of diabetes mellitus is estab-
insulin infusion is usually continued until urine lished, the chronic care phase begins. While the
ketone bodies are absent, which is often past the physiology is well understood and excellent treat-
persistence of hyperglycemia. This stage usually is ment is available, success in chronic daily care still
reached at the time that the child will be able to may be difficult. Successful control of diabetes in
tolerate oral feedings. This is the time to consider the younger child requires an attentive family
switching to subcutaneous insulin administration, which is able to deal with the calculations neces-
but the switch must be coordinated or a delay in sary in determining carbohydrate content of food
administering subcutaneous insulin after discon- and insulin dosage. Parenting style becomes para-
tinuing the i.v. infusion will allow the blood sugar mount. In the soon-to-be-independent teenager the
to rise again to dangerous levels. cooperation of the patient with the treatment plan
Close monitoring of mental status, vital signs, becomes the central focus. Families and patients
peripheral perfusion, and serum sodium and vary widely and the treating team must have expe-
potassium is important through the course. rience in dealing with the psychological overlay of
Evaluation of magnesium and phosphorus is also daily care of this chronic condition.
indicated initially, and if abnormal, at intervals It is important to impress upon the child and
thereafter (magnesium deficiency may cause family that the child should no longer be consid-
potassium loss in the urine). All the abnormal ered “sick” once a stable biochemical milieu has
measurements should improve in a continuous been established. Parents must be told that the
pattern; if sodium decreases rather than increases, condition is not preventable and that it is “not
excess free water administration may be the their fault” by their parenting nor their genes. The
cause; falling serum sodium is a risk factor for well-treated child will have few limitations on
the development of cerebral edema. their daily life and can look forward to a long life
Improvement should be continuous. Thus span. With poor care the opposite may be true.
deteriorating status may be due to worsening The state-of-the-art care for diabetes in child-
or unresolved acidosis, uncorrected or devel- hood and adolescence is provided by a team of
oping hypokalemia, hypoglycemia, hypoten- providers including diabetes nurse educators,
sion, untreated infection, or CE. dietitians, social workers and psychologists, as
well as the treating physician. Such a team may
Onset Without DKA not be available in smaller distant locations, but
With increased education of the population, some satellite clinics are often available at a distance
patients are diagnosed at earlier stages of the dis- from university centers or children’s hospitals.
ease and do not have DKA. BS might be only in Likewise telemedicine treatment can be provided
the range of 200–300 mg per deciliter. These anywhere in the world if a receiving site is avail-
patients might be educated in the outpatient able that can connect with the transmitting team.
environment if adequate and trained staff is At initial presentation, the family will be
available to administer such education. This is distraught over the new lifelong diagnosis, and it
appropriate only if ketosis is absent or minimal is necessary to take clues from them as to when
280 11 Diabetes Mellitus

they can be receptive to education about this dis- Table 11.5 Carbohydrate content of selected common
foods
ease. After the resolution of hyperglycemia and
ketonemia/acidosis, the major reason for a patient Starches Amount Carb (g)
to remain in the hospital is lack of ability to carry Bread, white or wheat 1 slice (1 oz) 15
out BS monitoring, insulin administration, treat- Hamburger or hot dog bun 1 bun 20
ment of hypoglycemia, or dietary planning, and Bagel 1 (Deli style) 60–70
the key to these accomplishments is family edu- Doughnut 1 medium 25
cation toward the understanding of these “sur- Pancake or waffle, frozen 1 small 15
vival” skills. In those families that cannot accept Tortilla, corn, 6″ 1.2 oz, each 10
teaching nor understand the teaching provided to Saltine 1 cracker 2
them, a lengthened hospitalization will result. It Oatmeal 1 cup (cooked) 30
Rice, white or brown 1 cup (cooked) 45
is important to involve the patient and family in
Spaghetti 1 cup (cooked) 40
the process of monitoring BS by home glucome-
Corn ½ cup 15
ter and the measurement and administration of
Potatoes
insulin as soon as they are able to accept teaching
Plain, baked Small (3 oz) 15
in the difficult time after the diagnosis. This early
French fries
education may shorten the hospital stay.
McDonald’s Small 25
Refried beans 1 cup 40
Apple
Dietary Management Small 4 oz 15
Medium 6 oz 25
The patient and family must understand appro- Large 8 oz 35
priate dietary management of diabetes, although Grapes 15 pieces 15
it will take some time before this becomes second Strawberries 1 cup (sliced) 10
nature. A dietician is expert in teaching these Apple juice 4 oz (½ cup) 15
skills and should be consulted early in the educa- Whole, low fat or skim 8 oz (1 cup) 12
tional process. The dietary treatment of children Yogurt (plain) 8 oz (1 cup) 12–14
with type 1 diabetes does not limit specific items Table sugar 1 Tbsp 15
as long as the insulin administered is appropriate Ketchup 1 Tbsp/1 pkt 5
for the carbohydrate load. However, an American Spaghetti sauce ½ cup 10
Diabetes Association (ADA) diet provides New York-style pizza 1 slice 45
healthy guidelines as a goal for the diet and con- Note: Exerpts accessed from the website of the Naomi
sists of 45–60 % of total calories from carbohy- Berrie Diabetes Center of Columbia University Medical
drates, 30 % from fat with less than 10 % Center (http://www.nbdiabetes.org/diabetes-faqs)
saturated fat, and 10–20 % calories from protein.
Birthday cake is not to be a staple of the diet, but
on occasion, it can be eaten with insulin cover- Table 11.5 contains a few examples of carbohy-
age, especially in the MDI regimen below. Total drate content of common foods for basic carbo-
recommended calories vary by age and size of hydrate counting.
the child. The family must learn carbohydrate
counting, a skill that will be especially important
if MDI or insulin pump is considered. Type 2 Preparation for Discharge
diabetes does require a carbohydrate-controlled
diet avoiding concentrated carbohydrates and The basic “survival skills” are learned before dis-
this difference between T1DM and T2DM may charge from the initial hospitalization so that the
become an issue in the family when relatives with patient can measure BS by glucometer, measure
type 2 diabetes try to apply their dietary approach insulin from the bottle or pen, administer the
to diet to children with type 1 diabetes mellitus. injection, use emergency measures including
Type 1 Diabetes Mellitus 281

intramuscular glucagon injection for hypoglyce- ries do not replace the maintenance of a
mia, and choose a reasonable diet. The patient meticulous record book, which assists the
must recognize the signs of hypoglycemia or patient and physician’s review of BS control.
impending DKA and the steps necessary to reach Most glucometer memories can be downloaded
the doctor for advice (not an easy task in a large to a computer or uploaded to web-based programs
medical center) and understand that therapy will for the generation of BS versus time-of-day
change if an intercurrent illness occurs. Although graphs and other useful analytic information.
the basics of adjustment of insulin may be taught These can be used in the clinic or e-mailed to the
in the hospital, in this age of mandatory rapid provider in between clinic visits. Nonetheless, all
discharges (mandated by funding agencies rather too often, the family brings in a glucometer that
than medical judgment), usually inadequate time was not set to the correct day and time and there-
is available to teach this all in detail and families fore cannot be interpreted, or they forget the glu-
can be overwhelmed with too much information cometer completely, leading to a wasted visit. If
too soon. Thus after discharge, the family/patient they keep a record book, they are involved in the
will call the physician daily for a few days or process of monitoring and have a second source
weeks to discuss the correct dose of insulin and of historical blood glucose data. Although glu-
explain why it was adjusted as such. This educa- cometers may have memory of the measurements
tional process aids the family in achieving inde- over the previous month or more, observation of
pendence in understanding the therapy of the patterns of BS changes is essential if a patient
diabetes. In time the family and patient who have is to learn how to make adjustments of insulin
observed the response to therapy will offer sug- doses and to guard against a dead battery falsify-
gestions for management that may be supported ing or emptying the glucometer’s memory.
by the team when deciding on change in therapy. Another reason to keep a written record is that
Hospitalization does not have to continue until some patients use multiple glucometers or may
blood sugar control is optimal. Because the not care for the device correctly: if the glucome-
patient will be more active at home than in the ter does not have the date or time set (perhaps
hospital and glucose values will therefore likely because of battery change or dropping the
decrease from values seen in the hospital, BS values device), the memory on the glucometer will be
do not have to be completely normalized before useless, as it will not be possible to determine
discharge. It is not necessary to expect the family when any glucose value occurred.
and child to have a complete understanding of the Families may believe that the child’s report of
biology of diabetes before discharge. An under- the blood sugar is “okay” when in fact it either
standing of the more complex details of the disor- has not been taken or is abnormally high or low.
der will develop during the month that follows The parents must see the actual numbers in the
the outpatient-management phase. glucometer on a regular basis to avoid a tendency
of many pediatric patients to “forget” to measure
blood sugar and report otherwise. Sometimes
Home Measurement by Glucometer pediatric patients will falsify blood sugar mea-
surements by using control solution rather than
No matter which schema is followed, careful their own blood. The patient may lick their finger
attention to BS monitoring is essential, at least before testing which factitiously lowers blood
before breakfast, lunch, and dinner, and before sugars as well. Others, usually teenagers, may
the bedtime snack. Home measurement of BS measure their blood sugar for a week or 2 before
by glucometer has revolutionized the ability of the outpatient visit having rarely done so in the
family and patient to control BS closely. weeks and months before the visit in an attempt
Many different glucometers are available for to show compliance. It is important to look at a
measurement of BS from drops of blood from the long history of blood sugar management mea-
finger, and all have various advantages and draw- surement rather than just the last 2 weeks before
backs. Most have memories, but electronic memo- the visit.
282 11 Diabetes Mellitus

Some glucometers are able to test blood acting insulin must be given before discontinuing
ketone levels. This is a more sensitive indication i.v. insulin to allow it to exert its effect while the
of impending ketosis and is be helpful in i.v. insulin effect wanes. If the child has been
management. receiving i.v. insulin for several days and the BS
has stabilized on oral intake, the total sum of
insulin that was infused over the previous 24 h is
Subcutaneous Insulin Management used as an initial estimate of the dose of insulin to
be given by subcutaneous injections over the next
Once BS is brought close to normal values, aci- 24 h. The total daily dose of insulin tends to
dosis is resolved, and the patient is able to toler- average between 0.5 and 1 U/kg/day; usually the
ate oral feedings, it is time to switch to lower range is used at first for safety, although
subcutaneous insulin. Many approaches are used significant variations are found to this number as
for insulin therapy, and more than one type of the course of the disease advances.
insulin and type of device are available for
administration (Fig. 11.2, Table 11.6). When the Two Dose Per Day Regimen
i.v. insulin is discontinued, the insulin effect will Historically management has involved two injec-
be quickly eliminated. Thus subcutaneous short- tions per day, each consisting of a moderate-

Three Methods of Administration of Subcutaneous Insulin

Multiple
Dose
Injections
Relative Action of Insulin

Ly Ly Ly Ly Ly Gl

3 Doses
per day

R+
NPH R NPH

2 Doses
per day

12 8 12 6 10 12
AM R+ AM PM R+ PM PM AM
NPH NPH
7:30AM 9:45AM 3:30PM 9:30PM
Breakfast Snack Lunch Snack Dinner Snack

Time of Day

Fig. 11.2 Three methods of administration of subcutane- time of action, R regular insulin although fast acting insu-
ous insulin. The curves represent the usual period of action lin might be used instead of regular in the 2 or 3 doses per
of each insulin preparation with the more heavily shaded day schemas, Gl glargine insulin (the action of glargine is
“tail” demonstrating the variable time until activity ceases. indicated by the light grey area on the top panel)
Ly lysproinsulin although insulin aspartate is similar in
Type 1 Diabetes Mellitus 283

Table 11.6 Actions of commercially available insulin morning and a dose of very-short-acting insulin,
preparations
lispro, glulisine or aspartate insulin, given before
Onset Peak Duration every meal or substantial snack by using a set
Preparation of action activity of action (h)
ratio of insulin-to-carbohydrate intake (e.g.,
Aspart 10–20 min 40–50 min 3 (to 5)
0.5–2 U insulin per 15 g of carbohydrate). The
Lispro 15–30 min 30–90 min 3 (to 5)
long-acting insulin may be considered to be the
Apidra 20–30 min 30–90 min 1–2.5
insulin that covers the needs of a patient who is
Regular 30–60 min 2–5 h 5–8
not eating and having stable activity per day; thus
NPH 1–2 h 4–12 h 18–24
it is insulin which covers the requirements for
Glargine 1h None 24
simple existence. In a noncompliant patient who
Detemir 1–2 h 6–8 h 24
misses insulin injections if they at least take one
long-acting insulin injection per day, the likeli-
hood of admission for DKA is considerably
acting (e.g., NPH) and a short-acting (e.g., decreased even if they missed many short-acting
aspartate insulin) insulin preparation. The insulin insulin injections. The rapid-acting insulin
dose may be estimated as two-thirds of total daily attends to the carbohydrates ingested and cor-
dose before breakfast and one-third of the total rects abnormal blood sugar values, an insulin that
daily dose of insulin given before dinner, with treats all the variation in living that occurs daily.
one-third of each dose given as rapid-acting and A dietician is essential in calculating this ratio of
two-thirds given as NPH insulin. Two injections insulin-to-carbohydrate intake. The patient or
per day management is a compromise, as the child family must count carbohydrates in each meal
is obliged to eat a meal containing a predictable consumed, so education is complex for MDI. The
carbohydrate content on a strict schedule to main- short-acting insulin is adjusted both on the basis
tain BS while the short- and moderate-acting of the carbohydrates to be consumed (determined
insulins exert their effects; skipping a meal will by carbohydrate counting) and the BS at the time
cause hypoglycemia as no carbohydrate peak of injection (determined by glucometer)
occurs at the time of the expected peak of the (Table 11.7). When the blood sugar is elevated or
action of the moderately acting insulin already decreased compared to normal, the insulin dose
given, whereas excessive carbohydrate intake is raised or lowered by a mathematical calcula-
will cause hyperglycemia. Thus two injections tion noted below. Someone (if the child is too
per day regimens are to be avoided if at all pos- young, some responsible adult) must be able to
sible. Some families are not capable or willing to calculate insulin doses at midday or at snack time
give a third noon injection or a fourth bedtime and administer the insulin while the child is away
injection of long-acting insulin, and the two dose from home. In this regimen, the child has a more
per day regimen is sometimes reluctantly accepted. flexible schedule, whereby meals might be
There are premixed insulins with 30 % short- delayed, extra snacks might be more easily
acting and 70 % moderate-acting insulin in the vial added, or meals may even be missed (this, of
or pen cartridge, but these are hard to adjust appro- course, is not a recommended action). Short-
priately in a child. Multiple-dose injection is a acting insulin may be given by the classic syringe
better schema for all children when possible. and needle with a bottle of insulin or by a more
convenient cartridge-and-pen system in which
Multiple Dose Injection Regimen the dose is dialed in and the insulin administered.
A much improved technique is the multiple dose Insulin vials or pens are usually kept at room
injection (MDI) regimen, sometimes known as temperature, where they are stable for 30 days.
basal bolus regimen, which involves the admin- The first step in preparing an individual who
istration of a long-acting insulin (glargine or was treated with two doses a day to multiple-dose
detemir insulin, which exerts a stable effect over injection is to determine the total insulin
the 24 h after an injection) every evening or requirements that the child has been using for the
284 11 Diabetes Mellitus

Table 11.7 Typical algorithm for short-term insulin cor- sleep period (BS should be measured at 3 a.m. on
rection (sliding scale)
a couple of occasions to be sure), the a.m. BS
For an average child over 10 years value will generally determine whether the dose
qAC of long-acting insulin is appropriate. If the BS
Blood glucose 151–200 Add 1 U Novolog stays stable between a normal premeal determi-
201–250 Add 2 U nation and the value 2 h later, the short-acting
251–300 Add 3 U insulin dose per a given serving of carbohydrates
301–350 Add 4 U is appropriate. If the premeal BS is too high, but
>350 Add 5 U and the calculated dose of additional short-acting
measure urinary
ketone insulin is able to decrease the BS value 2 h later
qHS to a desirable level, the sliding-scale dosage to
Blood glucose 201–250 Add 1 U Novolog control elevated blood sugars is appropriate.
251–300 Add 2 U Insulin is injected subcutaneously and is usu-
301–350 Add 3 U ally injected in the triceps area, the thighs, the
>350 Add 4 U and abdomen, or the lateral, upper buttocks and the
measure urinary injection sites must be changed regularly, or lipo-
ketone hypertrophy may develop, a “lump” under the
For an average child 4–10 years skin that will make the time of absorption vary
qAC from injection to injection and lead to unstable
Blood glucose 151–200 Add ½ U control. More rarely, insulin injections may lead
Novolog
to lipoatrophy and a depression occurs near the
201–250 Add 1 U
injection sites; cromolyn sodium ointment some-
251–300 Add 1.5 U
times is used to resolve this complication.
301–350 Add 2 U
>350 Add 2.5 U and
Pattern adjustments are taught to the family
measure urinary and child to allow them to respond to abnormal
ketones BS values with a change in insulin administra-
qHS tion. Thus they are instructed that the long-acting
Blood glucose 201–250 Add ½ U insulin should remain stable unless a need for
Novolog changing the dose is demonstrated by an aberrant
251–300 Add 1 U pattern of BS over at least 3–5 days. In contrast,
301–350 Add 1.5 U they are taught to change the short-acting insulin
>350 Add 2 U and at the time the injection is given, based on the
measure urinary
ketones concurrent BS.
These are starting doses that must be evaluated in each Thus depending on size and insulin sensitiv-
child after careful monitoring ity, one regimen might be to add 0.5 U starting at
a blood sugar of 151 mg/dL and add another
preceding 24 h. Approximately 40 % of that will 0.5 U for every subsequent elevation of 50 mg/
be long-acting insulin and the rest will be divided dL: e.g., 0.5 U of regular or short-acting insulin
into doses given before meals. A correction fac- to the basal planned dose if BS is greater than
tor is used for two dose per day injection that can 151, 1 U if BS is greater than 201, and 1.5 U if
be retained from the earlier days when it was BS is greater than 251 (Table 11.7). Older and
used for stabilization. All doses are modified larger subjects might get twice these doses, and
with observation of the child with time. empiric adjustment is used to find the best sliding
scale. The rule of 1,500 (some suggest to use
Time of Action of Insulin on Multiple Dose 1,800 when using the most rapid-acting insulin)
Injection Scheme (Tables 11.6) is useful to determine an appropriate sliding
If the patient is using long-acting insulin at scale: divide the total daily dose of insulin on a
bedtime, the BS at bed time is in the normal normal day into 1,500 (or 1,800), and the result is
range, and no hypoglycemia is noted during the the change in BS that might be expected with the
Type 1 Diabetes Mellitus 285

Table 11.8 General guidelines for the adjustment of daily insulin dose (for an average 10-year-old). The chart shows
the last 24-hour history of an example patient
Before breakfast Before lunch Before dinner Bedtime
Blood sugar (mg/dL) 240 260 160 150

Planned fast-acting insulin dose 10 10 10 –


Additional fast-acting insulin 2 3 1 –
on the sliding scale
Long-acting insulin dose – – – 30
(e.g., glargine)
In this case, the 10 U of fast-acting insulin plus 2 U on the sliding scale were not able to cover breakfast and bring down
the prelunch value, but the sugar stayed stable at ~250 mg/dL, so the 12 total units can hold the sugar values and probably
is the correct dose before breakfast with the addition of a sliding scale dose; if the a.m. blood sugar was normal, the lunch
value would likely be as well. Further, because the prebreakfast blood sugar was 240 and the bedtime glargine was 150,
it appears that the nighttime glargine dose was too low. If the pattern is seen over several days and if the blood sugar tested
at 3 a.m. is high, it is safe to add an extra unit or two of glargine insulin to the p.m. dose to bring down the prebreakfast
blood sugar. However, if the 3 a.m. blood sugar is low, the child is experiencing the Somogyi effect which raises the
morning blood sugar; in such a case, without the 3 a.m. value the prebreakfast value will falsely suggest that the nighttime
glargine dose is inadequate. If the breakfast blood sugar is brought down to 100–150 mg/dL by correctly adjusting the
glargine dose, the lunch value may very well follow suit with the new a.m. 12-U dose of fast-acting insulin. Usually it is
best to make just one change at a time and evaluate the results of each change. Although the sliding scale of fast-acting
insulin is designed to be given for changes in blood sugar encountered on a daily basis, a change in the premeal dose of
fast-acting insulin or the dose of glargine is usually not invoked unless a pattern is noted over several days. The adjust-
ments of rapid-acting insulin as described are usually applicable to adjustments in “regular” insulin as well

addition of 1 U of short-acting insulin. Thus if a ment regimen is compromising lifestyle. In gen-


child uses 50 U/day, the addition of 1 U as a slid- eral, a patient must be well prepared for the use of
ing scale should reduce the blood glucose level an insulin pump to avoid problems noted below.
by 30. One sliding scale may have to be devel- An insulin pump is computer controlled, small
oped for one meal, and another sliding scale for (the size of a pager), and contains enough insulin
another meal. Glucose correction at bedtime usu- for 3 days of infusion. It uses short-acting insulin
ally starts at a higher level, usually above 201 mg/ which through continuous subcutaneous infusion
dL, to make hypoglycemia during sleep less replaces the long-acting insulin injection of
likely while all insulin correction during the day MDI. The pump does not determine the infusion
usually occurs at a blood sugar above 151 mg/ rate nor the amount of a bolus of insulin to be
dL. All doses are modified with observation of given at mealtimes but rather the pump functions
the child with time. completely on the understanding of diabetes by
the patient or family and significant adjustments
Computerized Subcutaneous are necessary. The insulin pump must be pro-
Insulin Pump grammed by the family after careful attention to
The computerized continuous subcutaneous insu- blood sugars over a portion of the day and the set-
lin infusion pump (“insulin pump”) has improved tings changed appropriately. This process must be
diabetic management greatly. More and more repeated several times a year and must be adjusted
patients are being treated by an insulin pump soon as the child grows or enters puberty and insulin
after diagnosis. There are recommendations for resistance increases. One major advantage of a
an insulin pump to be invoked when hemoglobin continuous subcutaneous insulin infusion pump is
A1c is high, when there is frequent hypoglycemia the flexible basal infusion rates. Although a single
and the blood sugar values are quite variable, injection of glargine or detemir insulin lasts for
when microvascular complications or risks for 24 h exerting a rather stable effect, there is no
microvascular complications arise, or if the treat- way to vary its potency throughout the day.
286 11 Diabetes Mellitus

As described below there are variations in basal patient becoming aware of this if blood sugar
insulin requirements over a 24-h period; for monitoring is not frequent. If the tubing is kinked
example, the dawn phenomenon described below or clogged or exits the skin, the insulin infusion
leads to requirement for increased insulin before may stop unexpectedly. Short-acting insulin is so
awakening. A single dose of long-acting insulin quickly cleared from the body that severe insu-
cannot accommodate such changes. Insulin linopenia and even DKA will develop within
pumps can change the insulin infusion rate by hours if the user does not note such problems;
half-hour intervals, leading to many possible with multiple injection schedules the long-acting
changes per day on a preprogrammed schedule. insulin lasts for at least 24 h and offers a longer
Most patients use only three to five basal infusion margin of safety that cannot be accomplished by
rates per day, but the numerous possible settings the short-acting insulin pump infusion. The pump
may be used to improve control. The patient has alarms but cannot detect all problems that
administers a bolus of short-acting insulin at may arise. Thus attention to detail and frequent
mealtimes through the pump replacing the short- BS measurement are even more essential in pump
acting injection of MDI, depending on blood glu- therapy than in other forms of treatment. We
cose values and carbohydrate intake, by ordering believe that difficulties in the use of the insulin
the dose from a touch-pad keyboard on the pump pump might be avoided by initially testing the
or by a remote control. The pump has the ability ability of the patient to use it correctly.
to infuse a peak bolus, similar to a subcutaneous Continuous glucose monitoring is now avail-
injection of insulin, but has advantage of the addi- able and can be carried out for a period of several
tional ability to administer a square or dual wave days by using a tiny sensor that is temporarily
of insulin over a longer duration which will help implanted in subcutaneous tissue; this process
to control blood sugar which will tend to stay will alert users to trends in blood sugar control
elevated when fat or protein included in the meal and impending high or low values serving as an
(e.g., Pizza). Likewise small children may not alarm to dangers. The information can be used to
always finish their planned meals, and in this case analyze problems of glucose control or to deter-
half of the expected dose of insulin may be admin- mine pattern adjustments to insulin pump ther-
istered just before eating and the rest of the dose apy. However, the patient cannot use the glucose
adjusted to the amount of carbohydrate taken in reading on the monitor to administer insulin: a
given soon after eating. This method, like MDI, finger stick glucometer is more accurate and must
allows a more flexible schedule of meals and may be used for this purpose. At present one insulin
be especially attractive to teenagers. No pump can pump connected to its continuous glucose moni-
decide on the correct dose of insulin itself tor is able to suspend infusion when the monitor
(although promising research is aimed at produc- senses a significant drop in glucose. Another
ing a pump that can do just that). device is available that can transmit the blood
There are many provisos to successful insulin sugar value determined by the monitor at a dis-
pump therapy and serious dangers can arise in tance to a receiver that might, for example, be
pump therapy if the child and family are unable placed next to a parent’s bed should they be con-
to utilize it correctly. The family or child must be cerned about middle-of-the-night hypoglycemia
able to count the carbohydrate content of food in a child in another room.
and be able to enter it into the pump’s computer In all regimens, BS is monitored at least four
which will calculate the correct dose of insulin times per day (before meals and at bedtime,
for meal. They must scrupulously monitor BS always before eating), and the insulin dose is
values four times or more per day or control will adjusted according to these values. It is important
decline and offer no advantage over injections of to wait for 2–3 h after any preceding meal or
insulin. The infusion set (subcutaneous plastic snack to obtain a glucometer reading, as it takes
tubing) must be changed every 2–3 days, or it that long to overcome the inevitable peak resulting
will clog or even become infected, without the from carbohydrate intake. If the patient treats a
Type 1 Diabetes Mellitus 287

high blood sugar value with insulin less than period of increased activity without a change in
2–3 h postprandially, hypoglycemia may result insulin dose; this might be used to good advan-
due to the transient nature of the peak of glucose tage if unplanned activity occurs after insulin
measured following a meal. Insulin is not rou- has already been given. In general, blood sugar
tinely given outside of the scheduled time unless drops during exercise and exercise can be a good
the BS is measured very high at alternative times treatment if BS is slightly to moderately ele-
of the day (e.g., greater than 350) and ketone vated, but if BS is higher than 350 mg/dL, and
bodies appear in the urine. especially if ketones are present, exercise should
Initial settings for the insulin infusion rate be limited or curtailed completely until blood
are set and then adjusted based upon the needs sugar control is gained by insulin administration.
of the child after monitoring the results on blood Exercise during severe hyperglycemia can pre-
sugar. The basal infusion rate might be set cipitate ketonemia and ketonuria. Some children
according to the long-acting insulin the child will increase their BS with exercise, so values
was receiving for a 24-h day and dividing that must be checked before, during, and after exer-
by 24 or set the pump to deliver 1/24th of the cise while determining an individual child’s
child’s dose of long-acting insulin each hour. If response.
the child has not been on long-acting insulin,
calculate the dose as roughly 40 % of the child’s
weight. The dose of short-acting insulin for each
meal would be the same setting in the pump, as Glycosylated Hemoglobin
the child has been using previously for subcuta- or Hemoglobin A1c (Table 11.3)
neous insulin before meals. If the child has not
been using subcutaneous insulin prior to the ini- Glucose becomes nonenzymatically bound to
tiation of the pump, a rule of thumb might be a the amino terminus of the β chain of hemoglobin
dose of insulin of 1 U per 25 g of carbohydrate and measurement of hemoglobin A1c will indi-
for a young child, 1 per 15 g for the several cate the average blood sugar over the previous 3
years before puberty, and 1 per 10 g during months. The measurement reflects the last 12
puberty. The correction factor might be ½ U for weeks of average blood sugar levels with
every 50 mg/dL elevation starting at 151 mg/dL increased significance given to the values of the
blood sugar for young children and 1 U for previous 4 weeks. Because the glucose is revers-
every 50 mg/dL in older children as puberty ibly bound to hemoglobin for the week before,
develops. The correction might start at 201 the measurement that week plays no role in the
before bedtime. hemoglobin A1c value. The hemoglobin A1c
These guidelines are general, and individuals value is associated with an average blood sugar
do not follow a simple rule in their response to concentration. Current guidelines aim for a
calculated insulin dosage. For example, once hemoglobin A1c value under 7.5 for everyone
stabilized at home after discharge from the hos- under 19 years of age although this is considered
pital, when the child resumes the previous daily a target fully realizing that less than 25 % of
schedule, the insulin dose might be decreased patients under 19 years of age have reached such
(10 % or more) as the child will be more active a goal. In fact the target hemoglobin A1c for a
at home and may plan to participate in exercise, specific child may have to be modified based
as the activity will often decrease the blood glu- upon the circumstances and the abilities of the
cose value and could possibly precipitate hypo- family. “Tight control” leading to close to normal
glycemia; this might occur, for example, twice hemoglobin A1c values increases the risk of hypo-
per week, if the child has gym class on a twice- glycemia. Hypoglycemia may be less obvious in
a-week schedule. Alternatively, the family might younger children who may not report their symp-
have to find empirically the amount of carbohy- toms. Hypoglycemia may not even be noticed in
drate needed to maintain the BS during the those with hypoglycemic unresponsiveness.
288 11 Diabetes Mellitus

Other factors must be taken into consideration The Honeymoon Period


when measuring hemoglobin A1c. If hemoglobin-
opathies are being considered in a patient, a Most children with type 1 DM have a period of
hemoglobin electrophoresis should be carried out resumed insulin secretion and improved control
to determine if an abnormal hemoglobin is affect- as the pancreas recovers from glucose toxicity
ing the hemoglobin A1c value rather than actual caused by the extreme elevation of blood sugar.
blood sugar effects. Glycosylated albumin, fruc- This temporary recovery of pancreatic function is
tosamine, or total glycohemoglobin by affinity known as the “honeymoon period.” Some may
chromatography might be used as an alternative even not require insulin for months at a time; this
measurement of glucose control as these mea- period does not define a “cure,” but rather a tem-
sures directly relate to the hemoglobin A1c rising porary remission, and parents should not develop
with higher average blood sugar values but are false hope during this period that the disease pro-
not affected by abnormal hemoglobin. However, cess is over. Immunosuppressive therapy is under
fructosamine and glycosylated albumin reflect study as a means of prolonging the honeymoon
blood sugar control of the previous 1–2 weeks period.
rather than the 3 months of hemoglobin A1c. Beta cell function will decrease due to lipo-
Alternatively, 1,5-anhydroglucitol is a glucose- toxicity as well. This may occur only in the pres-
like monosaccharide found in food; intake is bal- ence of hyperglycemia. Lipotoxicity is usually
anced with excretion in the normal situation. discussed in terms of type 2 diabetes. However,
However, with hyperglycemia glucose blocks the lipid deposition in the beta cell is found in obe-
reabsorption of 1,5-anhydroglucitol so that val- sity and states of insulin resistance and impairs
ues vary inversely with blood sugar values and pancreatic function as well.
decrease with higher values of hemoglobin A1c;
values reflect control over the last 1 or 2 weeks
before the sample. Hemoglobin A1c can be mea- Sick Day Management
sured in minutes by office-based devices;
cross-checking the HgbA1c with the glucose mea- Sick day management refers to the changes neces-
surements on the glucometer and glucose values sary if there is a likelihood that the child’s intake
in the record book is a good way to evaluate the will vary from the normal level or if an intercur-
accuracy of the patient’s BS records or recollec- rent infection itself brings about a change in BS
tion of diabetes management. If, for example, a due to the stress of the illness; some children will
teenager has forgotten his or her glucometer and have an increase, but some will have a decrease in
written records at an office visit but reports that BS with infections and values will depend on oral
his or her blood sugar values are always about intake, so no hard and fast rule can be made. A
150 but hemoglobin A1c measured at the visit is change in insulin dosage will be determined by
over 14 indicating an exceptional degree of the regimen that the child uses for insulin admin-
hyperglycemia, a discussion should ensue istration. In a child taking multiple-dose injec-
between the patient and the parents; pediatric tions, the long-acting insulin dose will continue as
endocrinologist encounter such situations regu- it is necessary for basal maintenance of blood
larly. Thus, a HgbA1c value may lead to a frank sugar regardless of carbohydrate intake; for short-
discussion of reasons for poorer control than term illness, the long-acting insulin dose is usu-
expected and reported. It is important to recall ally not adjusted unless the illness is anticipated to
that the HgbA1c is an average, so that near-normal last for more than a day or 2 but may be reduced
levels are found with excellent control as well as 10 % for safety. The short-acting insulin dosage
with a mixture of high and low values of BS; a will be adjusted based upon carbohydrate intake,
near-normal value does not always indicate so even as the child eats less if the dose is adjusted
excellent control. appropriately, there should be no problem.
Type 1 Diabetes Mellitus 289

If the child however is on a two-dose per day The parents should be taught a general plan
regimen, the NPH insulin dosage is usually dis- to prepare for a sick day. Blood sugar must be
continued since this medication is given in antici- measured every 2–3 h and urine ketones at least
pation of a meal about 6 h later and during illness twice a day on a sick day. The child must be
such a meal may be skipped. Thus a child with kept well hydrated. Glucose-containing liquids
the two-dose per day regimen must have more may be offered to provide an opportunity to
frequent doses of short-acting insulin that substi- increase insulin dosage to cover the carbohy-
tutes for the NPH insulin. For patients on an insu- drates in the liquid and thereby provide another
lin pump, a temporary change in basal infusion means for decreasing ketosis. Unless the family
rate may be helpful if trends toward increasing or is well versed in sick day management, it is best
decreasing blood sugar values are noted since the to ask them to call the pediatric endocrine team
infusion rate is easily changed. if ketones in the urine are measured at more
For patients on insulin pump therapy, the than moderate or if blood ketones are rising. A
bolus injections will follow carbohydrate intake value for blood ketones under 0.6 mmol/L is
and should not present a problem during illness. normal, 0.6–1.5 nmol/L demonstrates rising
Testing for ketone bodies is essential while ketones which must be monitored further,
the child is ill as the symptoms of illness are 1.6–3 nmol/L presents a risk for diabetic keto-
much the same as the symptoms of ketosis. acidosis, and about 3 nmol/L indicates a serious
Testing for ketone bodies must occur whenever episode of DKA.
there is abdominal pain, nausea, and/or vomiting
regardless of blood sugar. If this step is elimi-
nated during illness or during an episode of nau- Hypoglycemia
sea or vomiting, impending DKA may proceed
unnoticed. Testing for urinary or blood ketone Hypoglycemia is generally defined as blood
bodies is to be used when blood sugar rises over sugar under 65 mg/dL, but diabetologists suggest
350 mg/dL. The presence of ketone bodies will using a cutoff of 70 mg/dL because it offers a
at the least indicate the need for increased hydra- margin of safety before symptoms occur. In fact
tion and will likely be an indication of the need recent guidelines suggest that any blood sugar
for increased or more frequent doses of insulin. low enough to cause significant symptoms might
Telephone contact is necessary with a provider be defined as hypoglycemia for the individual.
at the least during a sick day, if the family has no Symptoms of hypoglycemia can be adrenergic
experience in sick day management. If the such as sweating, shakiness, pounding heart beat,
patient cannot eat or drink, dehydration and sub- or neuroglycopenic such as stupor, irritability,
sequent DKA may develop and i.v. fluids become coma, or seizure. The episodes can be mild or
essential. If the illness involves nausea and vom- severe enough to cause death. Hypoglycemia will
iting and the patient cannot tolerate fluids, the occur in all individuals with DM at some point,
patient must come to the ED for evaluation; sev- and those with good control or those receiving
eral hours’ infusion of fluid may be all that is intensive therapy historically were more likely to
required, but hospital admission also may be have this complication because their general BS
necessary. It is not appropriate to give an anti- values will be closer to the low levels characteris-
emetic [e.g., promethazine (Phenergan)] to a tic of hypoglycemia; this may no longer be the
child with diabetes as an antiemetic may mask case with improved accuracy of glucometers
developing diabetic ketoacidosis; the vomiting and the availability of constant glucose monitor-
may be a sign of DKA rather than, for example, ing. Parents may be quite concerned about the
a viral illness with very different treatment occurrence of hypoglycemia and their children
required. Resolution of high BS and moderate and teenagers may be quite anxious about the
ketone bodies may decrease the tendency to risk for hypoglycemia. However, hyperglycemia
vomit, resolving some of the problem. is more damaging than mild hypoglycemia and
290 11 Diabetes Mellitus

appropriate control should still be encouraged. not responding to such measures and becomes
The sensation of low BS might be lost in those insensible, subcutaneous injection of glucagon
with chronically elevated BS values (hypoglyce- (0.02–0.03 or 0.5 mg/kg for children under 12 to
mic unresponsiveness). Further, more than half 1.0 mg or children over 12 years) must be given
of the episodes of hypoglycemia occur at night; to mobilize glycogen stores in diabetic subjects;
some children have night terrors as a sign of this the BS will increase considerably in a short time.
complication, but some have no evidence of Unfortunately, a glucagon injection may make
hypoglycemia or progress to seizures without the child nauseated and emesis will further
warning. Middle-of-the-night BS measurements complicate the day’s BS and insulin manage-
or a 3-day continuous glucose monitoring session ment. All individuals with diabetes must have
will help determine if this problem is occurring. easily available glucagon emergency kits that
Hypoglycemia can be extremely dangerous in contain the medication and a syringe and needle
certain circumstances such as participating in for administration. Intravenous 10 % dextrose in
extreme sports or driving, so preparations that saline at 2–3 mL/kg will provide emergency
will avoid hypoglycemia must be in place for treatment if all else fails.
such situations. No teenagers should be allowed Hypoglycemia may occur because of inappro-
to obtain a driver’s license if they are not care- priate insulin dosing. This may be due to basal or
fully monitoring their blood sugar and agree to bolus insulin dosage and the dose may have to be
test their glucose value before driving and agree changed if it was administered as instructed.
to stop the vehicle to retest and treat when symp- Inappropriate carbohydrate counting can also
toms occur. lead to hypoglycemia and further education
Treatment of hypoglycemia is accomplished should be invoked. Hypoglycemia may occur
by the administration of simple carbohydrates because of increased energy expenditure during
(not complex carbohydrates, which will raise the activity and the low blood sugar may not appear
blood sugar too slowly), but overtreatment may for 8–12 h rather than right after the activity.
lead to hyperglycemia and a seesaw effect of BS Education is important in the prevention of hypo-
values that are too high, leading to another dose glycemia. Anyone caring for the patient at home
of insulin that drives the value down, requiring or at school must know about the possibility of
another dose of glucose. Characteristically 15 g hypoglycemia and the treatment for it. A diabetes
of glucose is given to a child over 5 years of age, alert bracelet is essential to alert any care provider
and the blood sugar remeasured in 15 min but that the child has diabetes and is subject to
lower doses of glucose will be used in younger hypoglycemia.
children. If no rise in blood sugar is noted within Recurrent hypoglycemia may also be due to
10–15 min, another blood sugar is obtained after adrenal insufficiency. Type 1A diabetes mellitus
more carbohydrate is administered. A serving of may occur with autoimmune Addison’s disease
15 g of glucose is approximately the equivalent in autoimmune polyglandular syndrome number
of a 4 oz glass of juice or soda, two tablespoons 2 and possibly 3. Therefore evaluation for adre-
of raisins, one tablespoon of sugar, honey, or corn nal autoantibodies in patients with progressively
syrup, 8 oz of nonfat or 1 % milk, a few hard can- lowering insulin requirements and recurrent
dies, or three to four 4-g glucose tablets. Paste hypoglycemia is in order. Celiac disease may
glucose preparations are available as medical lead to fluctuating glucose levels as well.
preparations, but cake frosting might well substi-
tute if it is all that is available. These oral glucose
preparations may be absorbed through the buccal Somogyi Phenomenon
mucosa to a degree if the child is not cooperative
with swallowing. In 15 min, the BS should The Somogyi phenomenon occurs when hypo-
increase to more than 100 mg/dL, but if not, the glycemia triggers the release of counterregula-
whole process can be repeated. If the patient is tory hormones that subsequently increase BS;
Type 1 Diabetes Mellitus 291

thus a low BS at 3 a.m. may cause a rising BS in patient on long-acting subcutaneous insulin may
the morning. Nightmares, night sweats, and receive a 10–20 % decrease in the customary dose
headaches on awakening may occur in a subject during surgery. While the patient will be fasting,
with the Somogyi phenomenon. It would initially rapid-acting insulin will not be necessary, but
seem that more insulin is indicated at night insulin coverage of the intravenous glucose may
because the a.m. BS is high, but the opposite is be calculated at 1 U of insulin for every 4–6 g of
true. Increasing the p.m. dose of intermediate- or glucose administered. For 5 % dextrose there is
long-acting insulin or increasing overnight basal 5 mg of glucose per 100 mL. The goal is to
infusion from insulin pump would worsen the maintain blood sugar in the normal physiologic
situation, because the basic problem is too much range or slightly higher and to avoid ketosis.
insulin, leading to hypoglycemia. Management for a traumatic event might follow
the guidelines for management at surgery.

Dawn Phenomenon
Diabetes Team Management
The dawn phenomenon is an increase in blood
glucose in the early morning hours before wak- The American Diabetes Association and all
ing, due to an elevation of growth hormone and authoritative bodies agree that team management
ACTH/cortisol in the early hours. Alternatively is optimal for close control of diabetes at any age.
some children have an antidawn phenomenon in Thus a nurse educator, a dietician, and a social
which the increase in BS occurs just after mid- worker or psychologist, in the best of situations,
night rather than just before waking. Evaluation join the physician. This team approach may only
of these possibilities requires monitoring BS at be available at a larger medical center, which
intervals during the night. The 3-day continuous may be at a distance from the patient. Thus initial
BS monitoring can help evaluate these various education and quarterly consultations may be
patterns of blood glucose. offered, with day-to-day care provided by the
patient’s local physician. In some remote areas,
telemedicine consults are available from major
Surgery and Diabetes medical centers, so that advice might be dis-
pensed by two-way television connections if
If a child is to be kept NPO in preparation for and medical personnel are on site with the patient
during surgery, consideration of insulin manage- who can supply the necessary physical examina-
ment is essential. The best management during tions and provide the needed BS values over the
surgery would be the intravenous infusion of phone, fax, or monitor to the distant consultant.
insulin and glucose with constant monitoring of The aim for older patients is achievement of con-
blood sugar and changes in infusion rates as nec- trol of BS close to the normal range and achieve-
essary. If surgery is emergent and the patient is ment of a normal or near-normal value of HgbA1c,
not in metabolic balance, correction of the condi- but this is harder although desirable in younger
tion should be carried out as safety and the situa- children and infants. Adolescents and adults are
tion dictates. An infusion of 5 % dextrose with advised by the ADA to have (a) preprandial BS of
normal saline (there is controversy as to whether 80–120 mg/dL, (b) 2-h postprandial BS of less
normal saline or hypotonic fluids are best) along than 180 mg/dL, (c) bedtime BS of 100–140 mg/
with an i.v. infusion of insulin as is described in dL, and (d) HgbA1c of less than 7.5 %.
the management of diabetic ketoacidosis may be These values may be difficult to achieve and
carried out. If the procedure is short, a patient on are best used as goals rather than expected
an insulin pump may receive a customary night- accomplishments in all young patients. The goal
time infusion rate before surgery and a 10 % of a preprandial BS close to 150 mg/dL may be
decrease during the surgery. Alternatively a appropriate at younger ages to lessen the
292 11 Diabetes Mellitus

likelihood of hypoglycemia, fully realizing that control the blood pressure within 3–6 months.
even this relaxed goal might not be reached. Pharmacologic treatment is then considered if the
Considerable variation will be found in BS systolic and diastolic blood pressures are consis-
measurements in children of all ages because of tently above the 95th percentile for age, sex, and
the lack of precision of glucose management height or consistently over 130/80. If medication
with present technology, but variations should be is required, an angiotensin-converting enzyme
minimized as much as possible. (ACE) inhibitor is the first line of treatment for
Children are evaluated as needed for insulin diabetes. Every year, each child should have
adjustments and may receive advice over the determination of thyroid function (autoimmune
phone if they report or fax the BS values to the thyroiditis is common in T1DM and may occur at
physician, but these children should be seen in the presentation) by free T4 and thyroid-stimulating
medical office at least quarterly. Subjects’ heights hormone (TSH) if the initial determination at the
and weights are tracked to see if normal patterns time of diagnosis is negative. Fasting lipid deter-
are described. Decreased growth may indicate mination is performed after stabilization if the
poor control or the development of hypothyroid- child is in puberty or at 10 years if there is no fam-
ism or celiac disease. Decreased BS values and ily history of lipid disorders or at 2–8 years of age
decreased insulin requirements well after the time if there is a family history of significant lipid dis-
expected to span the honeymoon period might orders. If a fasting lipid panel is normal, it should
indicate the development of autoimmune be repeated in 5 years. If a fasting lipid profile is
Addison’s disease. Interphalangeal joints are eval- abnormal, it should be repeated annually to deter-
uated for contractures that develop after years of mine cholesterol and triglycerides (elevated lipid
DM. BP is monitored to determine whether values occur with poor control). If LDL choles-
hypertension has developed, possibly because of terol is over 130 mg/dL and there are risk factors,
kidney damage. All children should be examined and diabetes mellitus type 1 is a risk factor, or
for lipohypertrophy at the sites of insulin injec- over 160 mg/dL with no risk factors, current rec-
tion, as lack of rotation of sites may predispose to ommendations are to treat with statins (HMG-
this complication and lead to irregular absorption CoA reductase inhibitors), but it is essential to
of insulin. Lipoatrophy is rare with the use of remember that these medications are teratogenic
human insulin but was seen more often in the past and girls must not get pregnant while taking
with the use of animal insulin. Local reaction to statins. If the lipid level is not abnormal, rescreen-
insulin including erythema, swelling or edema, ing every 5 years is appropriate. Dilated compre-
warmth, or subcutaneous nodules sometimes hensive ophthalmologic examinations are
occurs at initial use which usually resolves. indicated annually to check for microvascular dis-
Although untreated type 1 DM leads to weight ease after the passage of 3–5 years with type 1
loss due to the loss of calories in the urine and cel- DM or at the onset of puberty or 10 years which-
lular starvation, treatment allows weight regain. ever is earlier and at diagnosis for type 2 diabetes
However, if a child consumes too many calories in older patients. A celiac reflex panel as evalua-
and increases insulin dosage to cover the intake, tion for celiac disease is indicated at diagnosis,
obesity may develop. Dietary counseling and especially if gastrointestinal symptoms, decrease
recalculation of insulin dose will help this prob- in growth rate, unexplained hypoglycemia, or
lem. Some teenagers may skip or decrease insulin deterioration of blood sugar control, are found
dose in a misguided attempt to lose weight, with since about 10 % of patients with T1DM have
sometimes tragic consequences. celiac disease. Vitamin D screening is also recom-
Every visit the child should have blood pressure mended in high-risk individuals. Neuropathy and
as well as height and weight and BMI determined. gastroparesis are rarely seen in the pediatric popu-
If blood pressure exceeds the 90th percentile for lation, but history and physical must be directed
age, sex, and height, dietary intervention with to evaluating these possibilities and sensation and
increased physical activity should be tried to proprioception should be checked even in the
Diabetes Team Management 293

young patient. Microalbumin-to-creatinine ratio manifestation of DM may wax and wane with the
determinations are carried out as the patient administration and cessation of the medication.
reaches the age of 10 years or the start of puberty, In some cases, the condition may become
whichever is earlier, or has had diabetes for 5 permanent.
years to evaluate the possibility of kidney damage Acquired forms of insulin-dependent DM in
in type 1 diabetes; this is carried out at diagnosis children with cystic fibrosis result from the degen-
in patients with type 2 diabetes. If elevated micro- eration of islet cell pancreatic function leading to
albumin is confirmed on two additional different loss of glucagon as well as insulin. Cystic fibrosis-
specimens from different days over in 6 months or related diabetes may appear in young patients and
if renal function testing shows disease, ACE with longer term survival in cystic fibrosis, the
inhibitor treatment may slow the progression of number of subjects with diabetes is increasing. In
renal disease, but these agents are teratogenic and this situation, maximal calories are required for
cannot be used in pubertal girls without an assur- nutrition which has implications for diabetes man-
ance of abstinence from sexual activity or the use agement. A team approach involving the pulmon-
of birth control. Thus uncontrolled diabetes is ologist, endocrinologist, clinical diabetes educator,
teratogenic as is treatment of hyperlipidemia and and dietitian is recommended. Evidence-based
renal disease; a teenage girl must always be coun- consensus statements point out that the diagnosis
seled on birth control or abstinence. is based upon a standard glucose tolerance test as
Dental care must be offered regularly because found in this chapter, although elevated fasting
of the risk of tooth decay or gum disease interfer- blood sugar or a 2-h postprandial blood sugar will
ing with the control of diabetes and nutrition. also suffice. Annual screening should occur after
Children with diabetes must be identified in 10 years of age. Standard self- or family-based
case they are separated from their guardians at a management as is customary in type 1 diabetes
time that they may be unresponsive. Although a should occur with blood sugar measured at least
bracelet or necklace can be obtained from most three times a day and hemoglobin A1c monitored
drugstores, the MedAlert system will keep track quarterly. Insulin is the only appropriate treatment
of basic medical information and physician’s in that the disease originates from the destruction
phone number and is preferable as a means of of the beta cells. Diabetic complications including
identification. nephropathy and dyslipidemia may occur and
Summer camps for children with diabetes are yearly measurement of microalbumin-to-
available nationally and are a wonderful way for creatinine ratio in urine should be carried out.
patients to meet others with the same problems Fasting lipids are measured annually if abnormal
and, aside from realizing that they are not unusual and every 5 years if normal.
in their condition, to learn ways of dealing with Chronic pancreatitis may lead to a picture
the disease. Local diabetic groups (lay or medi- condition similar to cystic fibrosis-related diabe-
cally based) offer support and information for the tes and monitoring should proceed as for T1DM.
maintenance of good metabolic control.
Long-Term Complications
Large national or international studies of individ-
Iatrogenic or Other Acquired Forms uals with diabetes to determine who is likely to
of Diabetes Mellitus Type 1 develop complications (e.g., the Diabetes Control
and Complication Trial, DCCT) provided invalu-
Various drugs can precipitate DM, particularly able information in determining improved or opti-
drugs used in transplantation (such as FK506) mal therapies for diabetes. These pivotal studies
and others used in therapy for cancer (e.g., provide evidence-based guidelines for therapy.
L-asparaginase or prednisone for acute lympho- When aspects of diabetes care can be determined
cytic leukemia), which complicate the manage- from such studies, treatment paradigm should
ment of those serious conditions. The clinical adhere to them in an evidence-based manner.
294 11 Diabetes Mellitus

Nationwide study (DCCT) and international with dilation or retinal photography is recom-
studies demonstrate a dramatic decrease in com- mended yearly as described on the schedule
plications of type 1 DM with “tight control,” above. Nephropathy may develop as puberty pro-
which involves maintaining the HgbA1c and BS gresses in a child with diabetes or with the
values close to normal; 99 % of the variation in increased duration of the disease. The glomeruli
risk was attributed to the level of hemoglobin A1c. may become sclerotic after initial thickening of
The DCCT also showed that poor control during the basal membrane, so urine evaluation for
adolescence leads to increased microvascular microalbumin by urinary microalbumin-to-
complications even 5–7 years later. Maintaining creatinine ratio must be carried out yearly after
blood sugar close to normal does increase the risk the onset of puberty. Improved control will delay
for hypoglycemic reactions, but this may be or eliminate this complication and even reverse
minimized by the use of modern monitoring such changes, but the use of ACE inhibitors will
techniques. The present American Diabetes help limit progression.
Association goal for control of diabetes is a hemo- Hypertension may develop in a patient with
globin A1c below 7.5 or any patient under 19 years diabetes usually who is in poor control.
of age. This is an evidence-based recommenda- Microalbuminuria might be the first indication of
tion based upon the fact that hyperglycemia will kidney damage which leads to hypertension or
increase the risk of early complications of diabe- elevated blood pressure may proceed to microal-
tes including heart disease and kidney disease. buminuria. Hypertension can lead to increased
Microvascular and macrovascular complica- risk for cardiovascular events and treatment with
tions of diabetes in general are related to the ACE inhibitor is generally suggested as the first-
duration of disease and degree of control. The route therapy. (Hypertension is mentioned in
DCCT, as noted earlier, demonstrated that tight Chap. 13 as well.)
control with BS close to normal will decrease the Neuritis is rare in the initial presentation of
development of these complications and even DM but has been reported. Neuropathy in the
reverse them. Long-term complications of diabe- extremities is usually but not always found in
tes are found in the capillary endothelial cells in adults with diabetes and long-term poor control.
the retina, mesangial cells in the renal glomeru- Macrovascular disease of the cardiac vessels is
lus, and neurons and Schwann cells in peripheral rare in childhood but will develop with poor con-
nerves. These cells cannot reduce the concentra- trol as the patient ages. This can also be fore-
tion of glucose within them as other cells in the stalled by good control. Lipid values increase
body and thus are exposed to high levels of glu- with poor control, and this factor, genetic tenden-
cose for a longer period than other cells. Damage cies, and the hypertension of kidney disease all
to the cells occurs through the polyol pathway. can increase the likelihood of such macrovascu-
When glucose concentrations within the cells are lar disease.
elevated, aldose reductase grows, which would Serious infections, including mucormycosis,
ordinarily reduce the presence of toxic aldehydes are most prevalent in patients with long-term
by conversion to inactive alcohols, in this case poor control.
reduction of glucose to sorbitol, which subse- Pancreatitis may lead to diabetes if pancreatic
quently is oxidized to fructose. The aldose reduc- function is severely affected, but conversely, ele-
tase consumes NADPH in this process, thereby vated lipid levels caused by DKA or very poor
eliminating the production of reduced glutathione, control may themselves lead to pancreatitis.
an important intracellular antioxidant, leaving The Mauriac syndrome describes patients
the cell exposed to oxidative stress. with poorly controlled diabetes mellitus who
Retinopathy develops through changes in the develop hepatomegaly and short stature.
retinal capillaries and may lead to visual impair- Improved care will reverse this situation, but
ment or blindness. Improved control will fore- there is a concern for progression of retinopathy
stall this complication. Thus ophthalmoscopy and complications.
Diabetes Team Management 295

Poor growth can occur in poorly controlled such as diabetes and over also 130 mg/dL in dia-
diabetes mellitus without the full Mauriac syn- betes with additional risk factor(s). Unfortunately
drome developing. However, celiac disease or the generally recommended medical treatment,
hypothyroidism may likewise lead to poor growth the use of statins (HMG-CoA reductase inhibi-
and must be eliminated as a cause. Poor control tors) as described in Chap. 13, has not been
will also lead to delayed puberty and irregular shown to lower future cardiovascular risk in dia-
menstrual periods or amenorrhea. betes in children and there are no long-term stud-
While no pediatric patient should be smoking ies of their effects in children with or without
(nor anyone else), individuals with diabetes suf- diabetes, so even the decision of starting treat-
fer an increased risk from this habit. Microvascular ment is controversial.
complications may increase with smoking and
may also lead to persistent micro- and microalbu- Transition to Adult Care
min area. There is some evidence that smoking The care of an adolescent with diabetes involves
may predispose to the development of type 2 increasing the patient’s increased responsibility
diabetes. for his or her own care and increasing indepen-
Likewise alcohol is a risk factor in adolescents dence with parents. This is a stressful time in
with diabetes which may predispose to hypogly- many families and conflicts arise as teenagers
cemia. If there is prolonged alcohol intake in the will not characteristically follow all care plans.
individual with decreased nutritional intake, the A nonjudgmental approach with open discussion
person is in effectively in the fasting state; is necessary and counseling sometimes must be
glycogen stores will be depleted which tends to invoked during the process.
lead to hypoglycemia. Further alcohol ingestion As reproductive maturity is reached in females
can interfere with gluconeogenesis since ethanol with any type of diabetes, another responsibility
is metabolized by alcohol dehydrogenase in the emerges. Diabetes in girls of reproductive age
liver to oxidize to acetaldehyde and acetate which brings up the questions of complications to
requires nicotinamide adenine dinucleotide mother and child during pregnancy. Untreated
(NAD) as a cofactor; this limits the abundance of diabetes is dangerous to a mother and to unborn
NAD causing reduced gluconeogenesis and aci- fetus. Poorly treated maternal diabetes may lead
dosis. Thus alcohol eliminates the second line of to the classic outcome of the infant of the diabetic
defense against hypoglycemia. mother with high birth weight and tendency to
Another complication of diabetes is a risk of hypoglycemia, alternatively a small-for-
impotence, a discussion that should be held with gestational-age infant who carries metabolic
noncompliant pubertal boys in hopes that this risk problems throughout the life or teratogenesis of
may motivate them toward better compliance. the heart and other organs. A macrosomic infant
Dyslipidemia is common in the presentation of a diabetic mother has a risk of shoulder dysto-
of diabetes. After appropriate control, lipid levels cia during delivery and the mother has risk for
should return to the baseline for that individual. undergoing caesarian section. Infants of diabetic
With poor control that may not occur. A heart- mothers may develop jaundice after birth or
healthy diet is always appropriate for someone hypocalcemia in addition to the likelihood of
with diabetes. Use of medication for hyperlipid- hypoglycemia and hypocalcemia.
emia requires specific guidelines and there is In a normal pregnancy there is an increase in
some flux in these guidelines. Treatment for hepatic gluconeogenesis which can be controlled
dyslipidemia is different in diabetes than in the by increased insulin secretion in a mother
average individual without a risk. Thus medical without diabetes but will not be as well controlled
treatment may be considered in a child over during gestational diabetes. The insulin sensitiv-
10 years with an LDL cholesterol level unre- ity of a pregnant woman who has progressive
sponsive to lifestyle change over 190 mg/dL development of insulin resistance due to gesta-
without risk factors, over 160 with a risk factor tional diabetes may decrease more than 50 %
296 11 Diabetes Mellitus

from the preconception state. Mothers also have nancy is appropriate. Careful maintenance of
a lower threshold for developing DKA than blood sugar is possible and is required during
before pregnancy. A mother with gestational dia- pregnancy for a healthy outcome of mother and
betes who has autoantibodies found also in type 1 child. Of course, institution of healthy manage-
diabetes may very well be developing type 1 dia- ment techniques for diabetes should occur many
betes. In the absence of antibodies a woman with years prior to the pregnancy.
gestational diabetes has a future risk for the The teenage years also involve other responsi-
development of type 2 diabetes and/or obesity. bilities of impending young adulthood. Driver’s
The mother with gestational diabetes carries a license will be sought but cannot be awarded to
risk for preeclampsia, hypertension, preterm someone who does not carefully monitor his or
delivery, miscarriages, stillbirth, perinatal death, her blood sugar to make driving safe.
and polyhydramnios. The child carries a risk of Hypoglycemia is well known to impair judgment
congenital malformations including congenital and will render the patient incapable of safely
heart disease such as tetralogy of Fallot, transpo- driving a car. In both cases if noncompliant teen-
sition of the great arteries, septal defects, and agers do not take care of their diabetes or do not
anomalous pulmonary venous return; central measure their blood sugars to allow them to treat
nervous system and other neural tube defects hypoglycemia or hyperglycemia appropriately,
including anencephaly, spina bifida, encephalo- they are not appropriate candidates for driving.
cele, and hydrocephaly; and abnormalities of the The driver with diabetes must always measure
ear including anotia/microtia, orofacial clefts, blood sugars before turning on the engine to
limb defects, and defects in the urogenital sys- ensure that BS is in target range. Long-range
tem. Sacral agenesis/caudal dysplasia is more drivers require retesting at intervals. There must
commonly found in infants of diabetic mothers be ready supply of glucose available for the driver
compared to the general population. Macrosomia, especially if they are along vehicle. Because of
small for gestational age, polycythemia, iron the constant risk for auto accidents with or with-
deficiency in developing organs, which may lead out diabetes the driver should have a MedAlert
to cardiomyopathy and altered neurodevelop- bracelet or necklace to indicate the presence of
ment, hypertension and cardiac hypertrophy due diabetes in the unfortunate situation should there
to catecholamine production, and impaired or be an injury rendering the person incapable of
delayed lung maturation may occur. The infant of communicating this fact.
a diabetic mother also has a lifelong increased It must be emphasized to the family in patients
risk for obesity and type 2 diabetes. Thus coun- who are not complying that there is a significant
seling all reproductive-age girls in contraceptive ethical risk to an irresponsible teenager driving
techniques or abstention is essential. with the risk of hypoglycemia and that if stopped
Preconceptional counseling has been demon- by the authorities the teenager will be considered
strated to be effective in decreasing complica- “driving under the influence” which is not a sym-
tions and should include discussion with the pathetic reception from the police or Highway
mother of potential fetal and maternal complica- Patrol. If fatalities should occur during poorly
tions, adjusting medications to aim for a hemo- controlled diabetes, the charge could very well be
globin A1c below 6.5 in most cases and vehicular manslaughter. These unpleasant
emphasizing the importance of ongoing follow- thoughts must at least be discussed in some man-
up during pregnancy. Since early hyperglycemia ner to a noncompliant teenager whose parents
occurs at a time when fetal organs are first devel- plan to offer the car keys. It is the provider’s
oping, optimal control must be achieved before responsibility to determine if the teenager is not
pregnancy for optimal outcome. At the first dem- responsible enough to drive and if that is the case
onstration that a girl with diabetes is pregnant, to report to state licensing agencies as medical
transition to a high-risk management team or providers are mandatory reporters. This will lead
specialist in the care of diabetes during preg- to a long delay in regaining a license even if the
Neonatal Diabetes 297

teenager starts cooperating with diabetic control. 1 in 500,000 but more recently is considered to
Driver’s licenses in some states require answer- be a much more common condition. In some
ing the question “do you have a condition which families adults manifest other diagnoses such as
will impair your ability to drive the car.” Well- MODY or type 1 or type 2 diabetes while the
controlled diabetes might not seem to be such a children have NDM. Intrauterine growth restric-
condition but poorly controlled diabetes certainly tion and SGA presentation are common in neona-
qualifies. tal diabetes mellitus. Some forms of neonatal
As the teenage years pass it is important to diabetes can be treated with sulfonylureas after
prepare an adolescent with diabetes for the next initial stabilization with insulin which makes the
phase of care which will involve a provider who specific diagnosis important in determining
cares for adults as they leave the pediatric prac- management. The terminology generally divides
tice. The age of transition is dependent upon the the conditions into transient and permanent
patient and family and their wishes but will NDM. Not all authorities use this classification as
almost always occur by 21 years of age. The tran- some of the apparently transient conditions that
sition is to be planned often in writing and is best last for over 1 year are classified in one schema as
discussed years in advance. An ideal situation permanent NDM.
would be a clinic or environment where both
pediatric and adult providers practice side by side
but these are rare. An optimal situation will also Transient Neonatal Diabetes Mellitus
be the identification of the adult provider before
the transition occurs. However, the transition of Transient neonatal diabetes mellitus may last for
care is not always easy as providers for adults a few years or remit at puberty. Three genetic
may not accept the insurance plan of the patient. subtypes are noted. Type I transient neonatal dia-
Teenagers, according to present law, will stay on betes (#601410 ICD+DIABETES MELLITUS,
the parent’s private insurance plan until 26 years. TRANSIENT NEONATAL, 1) presents with
The care administered in a provider’s office for SGA and requires neonatal insulin therapy due to
an adult may not include the team-based approach nonketotic hyperglycemia which usually remits
which is optimal during childhood and adoles- by 12 weeks of age. Elevated blood sugar may be
cent diabetes care. Thus transition is not always noted during intercurrent illnesses thereafter and
easy process, hence the need to prepare for it long between 4 years of age and at puberty type 2 dia-
in advance. betes may develop. Type I is due to a mutation in
the imprinted, paternally expressed ZFP57 gene
which lies within the imprinted promoter of
Neonatal Diabetes the PLAGL1 gene on chromosome 6q22-q23.
Type II transient neonatal diabetes (#610374
Rarely DM is found in the neonate or child up to ICD+DIABETES MELLITUS, TRANSIENT
6 months of age without laboratory finding of an NEONATAL, 2) and type III transient
autoimmune basis of type 1 DM. This condition neonatal diabetes (#610582 ICD+DIABETES
has been called neonatal diabetes mellitus or MELLITUS, TRANSIENT NEONATAL, 3) are
monogenetic diabetes of infancy; for this discus- due to mutations of the KATP channel of the
sion we will call it neonatal diabetes mellitus, or β-cell membrane; mutation in the ABCC8 gene
NDM. Some patients with NDM present between of the Kir6.2 subunits is found in type II which is
6 and 12 months of age. Type 1A diabetes melli- found with heterozygous and homozygous inher-
tus is rare under 6 months of age. The etiology of itance and KCNJ11 of the SUR1 subunit is found
NDM is found in a family group of at least 20 in type III which is autosomal dominant. Both
genetic disorders. Neonatal diabetes was origi- type II and type III transient NDM respond to
nally reported with a prevalence of approximately oral sulfonylurea therapy.
298 11 Diabetes Mellitus

Permanent Neonatal Diabetes Type 2 Diabetes Mellitus


Mellitus
Type 2 DM is the preferred term for what was pre-
Permanent neonatal diabetes mellitus (#606176 viously called non-insulin-dependent DM (TYPE
ICD+DIABETES MELLITUS, PERMANENT 2 DIABETES MELLITUS #125853 DIABETES
NEONATAL; PNDM) may be due to either of MELLITUS, NON-INSULIN-DEPENDENT;
five mutations, but the most common three listed NIDDM variously at 20q12-q13.1, 20q12-q13.1,
below are heterozygous autosomal dominant 17q25, 13q34, 11p12-p11.2, 2q32, 2q24.1). Type
forms. Activating mutations in the subunits of the 2 diabetes is due to a combination of insulin resis-
ATP-sensitive potassium (KATP) channel of the tance and of impaired first-phase insulin secretion
β-cell membrane can be responsible for transient from the beta cell of the pancreas leading ulti-
or permanent neonatal diabetes mellitus. These mately to more severe insulinopenia. Type 2 DM
include the KCNJ11 gene of the Kir6.2 subunit was previously considered a disorder of adults,
which is more common in permanent neonatal but American Indian adolescents were reported to
diabetes mellitus or ABCC8 gene of the SUR1 develop this condition over 20 years ago. About
subunit which is more common in transient neo- the same time a tenfold increase in type 2 DM was
natal diabetes. Infants with the KCNJ11 mutation reported in African-American teenagers and chil-
may have developmental delay due to associated dren. Now, with the epidemic of childhood obe-
mutations in their neurons. Those with severe and sity across the USA and the associated insulin
developmental delay and epilepsy along with resistance, an increase in type 2 DM is found in all
neonatal diabetes mellitus are considered to racial/ethnic groups in the USA, but prevalence is
have the DEND syndrome. The diabetes in these highest in Native American, African-American,
conditions can usually be treated with Hispanic-American, and Asian-American teenag-
sulfonylureas. ers and children, and the condition is appearing
Mutations in the proinsulin gene (INS) also throughout the world in regions where the preva-
lead to permanent neonatal diabetes, but this lence of obesity is increasing. The prevalence of
form requires treatment with insulin and not type 2 diabetes in adolescence now exceeds the
sulfonylureas. prevalence of type 1 diabetes in American Indian
In addition to the autosomal dominant forms teens in the USA. It is more common than type 1
noted above, autosomal recessive forms are diabetes in children in Hong Kong, Taiwan, and
reported particularly in families with consan- Japan although children in Asia tend to have
guineous marriages. While heterozygous muta- lower BMI associated with the type 2 diabetes
tions in the glucokinase gene lead to MODY type than is found in the USA. This condition has a
III, homozygous loss-of-function mutations in stronger genetic pattern than type 1 DM so that
the gene for glucokinase lead to NDM with the there is usually a family history of type 2 diabetes.
need for lifelong insulin therapy. T2DM may be found in children as young as 6
Pancreatic agenesis has been related to autoso- years, although it is most prevalent in the teenage
mal recessive homozygous deficiency of PDX1, years when there is a normal physiologic increase
which is also the cause of MODY4 (#606392 in insulin resistance. In spite of the obesity epi-
ICD+MATURITY-ONSET DIABETES OF THE demic, type 2 diabetes in childhood and adoles-
YOUNG, TYPE 4; MODY4). cence is not epidemic, with a prevalence of 0.46
Genetic diagnosis is available for NDM and per 1,000, but certainly must be considered when
appears most cost-effective in infants presenting evaluating any child with obesity especially with
with diabetes under 6 months of age with the symptoms of diabetes.
findings of neonatal diabetes mellitus and no The physical finding of acanthosis nigricans,
pancreatic autoantibodies. an area of darkened and thickened skin found at
Type 2 Diabetes Mellitus 299

the back of the neck or in flexural creases, is some appear to have latent-onset diabetes of the
physical evidence of insulin resistance. The adult (LADA) or latent-onset diabetes of the
description of the later development of diseases young (LADY) which has an indolent course of
in individuals with impaired fetal growth or progression to the requirement for insulin ther-
infantile catch-up growth (the Barker hypothesis apy. Glucose toxicity occurs at the time of diag-
or the early life onset of adult disease; see Chap. nosis and in states of hyperglycemia impairs beta
5) includes an early development of insulin resis- cell function, but some recovery will occur after
tance and type 2 DM. Type 2 DM is found more euglycemia is established; thus type 1 DM is the
frequently in children and teenagers with large apparent initial diagnosis, but type 2 DM becomes
birth weights as well as those with decreased the favored diagnosis with the passage of time.
birth weights. Likewise in type 2 diabetes, since lipids are often
Unlike classic type 2 DM in adults, children extremely elevated at diagnosis, lipotoxicity of
can develop ketoacidosis presenting in a manner the beta cell also occurs, but recovery will follow
very similar to type 1 diabetes, confusing the dif- resolution or improvement of hyperlipidemia
ferential diagnosis between type 1 DM and type with normalizing blood sugars.
2 DM. Type 2 diabetes is an insulin-resistant con- The mainstay of therapy for type 2 DM should
dition and the features of type 2 diabetes are the be appropriate diet and exercise, and lifestyle
same as those of the metabolic syndrome. changes leading to weight loss which in some
Evidence-based guidelines were developed cases will accomplish better control than the use
for type 2 diabetes in pediatrics. Random screen- of oral medication such as metformin. Adherence
ing was not suggested, but when a child with obe- to the Academy of Nutrition and Dietetics’
sity is under evaluation for other comorbidities, PEDIATRIC WEIGHT MANAGEMENT
the search for type 2 diabetes is appropriate. (PWM) GUIDELINE is suggested by the AAP.
Certain laboratory tests are indicated at the time However, if the patient is suffering from acute
of diagnosis of type 2 diabetes and annually metabolic derangements, medical therapies for
thereafter; these include fasting lipid levels (if hyperglycemia are usually needed at first. If there
normal initial test screening every 5 years), liver is no DKA, and if the hemoglobin A1c is below 9,
enzyme determination, urinary microalbumin-to- metformin, a biguanide approved for children
creatinine ratio, ophthalmologic examination, older than 10 years, may be initiated. Metformin
and evaluation for obstructive sleep apnea. is generally considered safe but can precipitate
Evaluation for PCOS in girls with suggestive lactic acidosis, and a metabolic panel must be
medical history is also appropriate. Blood pres- determined to ensure normal function of liver
sure must be monitored carefully. and kidney before initiating therapy. In addition,
Diagnosis is made in the manner described rhabdomyolysis and elevated creatinine kinase
above for diabetes with elevated fasting blood may rarely occur. Since metformin may cause
sugars or 2-h postprandial blood sugars on sev- vitamin B12 (cyanocobalamin or cobalamin) or
eral measurements or sometimes in the absence folate deficiency, supplementation with these
of symptoms of obvious hyperglycemia but sus- vitamins is suggested. A dose of 500 mg is used
picion of T2DM by the glucose tolerance test as at first, and if adequate control is not achieved,
described above. The antibodies noted above that the dose may be increased by 500-mg increments
are found in type 1 diabetes should be measured at 1–2-weekly intervals up to a maximum of
in apparent type 2 diabetes as they may indicate a 2,000 mg divided into two doses per day in larger
true diagnosis of type 1 diabetes or the risk for children up to a maximum of 2,500 mg/day in
further autoimmune diseases. However, adults older teens. Abdominal distress may occur with
and children with T2DM may have the same anti- this therapy but often resolves within a few
bodies as in type 1 diabetes confusing the issue; weeks; the medication should be offered with
300 11 Diabetes Mellitus

meals. Metformin by itself cannot cause hypo- although as stated above they may be negative in
glycemia since the main action is to decrease some patients with type 1 DM as well. If the
hepatic glucose production but not to increase antibodies are positive and the patient otherwise
insulin secretion. However, if metformin has appears to have type 2 diabetes, the patient may
been used in a patient treated with insulin, insulin have a slowly progressive type 1 diabetes which
dosage is decreased as BS decreases with metfor- at least temporarily does not require insulin
min, as unintended hypoglycemia is a possibility treatment for a year or 2 (latent autoimmune dia-
when insulin responsiveness increases. betes in the young or LADY). Since exogenous
Insulin is the first line of therapy in a patient insulin is usually administered in the acute
with significant symptoms, if hemoglobin A1c is hyperglycemic phase of any type of diabetes,
over 9, if the blood sugar is ≥250 mg/dL, or in insulin assays will be inaccurate if the sample
whom the differential diagnosis between types 1 was taken after insulin therapy because of inter-
and 2 is unclear. If the hemoglobin A1c is higher ference in the assay by insulin itself or because
or symptoms are present after 4 months or if of antibodies that may develop against the
hemoglobin A1c does not descend to less than 7 administered insulin. Thus measurement of
on lifestyle or metformin therapy, insulin therapy serum C-peptide cannot clearly separate type 1
is initiated. Sometimes long-acting insulin will and type 2 diabetes at diagnosis but may be use-
be adequate for initial therapy, but multiple-dose ful after disease has been present for at least 1
injection may ultimately be indicated and may be year when type 1 diabetes patients cease to
required for the long term. Blood sugar moni- secrete insulin in most cases. Observing the
toring in the manner described above for type 1 course of the patient is necessary in frequent
diabetes is carried out in type 2 diabetes on cases before the diagnosis is determined.
insulin therapy as well. With successful weight It is not yet clear what the prognosis and com-
control, oral hypoglycemic agents and insulin plications of type 2 DM will be in young sub-
doses may be decreased or, in some cases, jects, but the development of complications
stopped altogether. appears to move faster than in younger adults
The differential diagnosis between type 1 dia- who have type 2 DM and who have serious and
betes in the honeymoon phase and type 2 diabe- early complications if control is poor. By exten-
tes can be difficult. Patients in the honeymoon sion, the complications of type 2 diabetes may be
phase of type 1 DM may be able to secrete more serious in those adolescents and children
endogenous insulin and C-peptide, suggesting who do not achieve adequate control.
that they have type 2 diabetes further clouding Type 2 diabetes might be found in Turner syn-
the differential diagnosis between type 1 and drome or Prader–Willi syndrome or any condition
type 2 DM. In the obesity epidemic one cannot prone to weight gain as described in Chap. 13.
automatically assume that a child with obesity Prevention of type 2 diabetes goes hand in
has type 2 rather than type 1 diabetes. Likewise hand with prevention of obesity, and the approach
the increased prevalence of type 2 diabetes in to this major public health problem is described
certain racial/ ethnic groups does not mean that in Chap. 13.
they cannot instead develop type 1 diabetes. Atypical diabetes mellitus (#612227
Further, the elevated blood glucose at the time of ICD+DIABETES MELLITUS, KETOSIS-
diagnosis may exert glucose toxicity on the beta PRONE; KPD) or ketosis-prone diabetes some-
cells, limiting their ability to secrete insulin to times known as Flatbush diabetes often presents
any appreciable degree, suggesting type 1 diabe- with ketoacidosis but after a short period of insu-
tes and making the differential diagnosis even lin treatment may go months to years without the
more unclear. Type 2 DM is not an autoimmune need for further insulin treatment. Some patients
condition, so anti-islet cell, anti-insulin, or anti- appear to have a heterozygous and some a homo-
GAD antibodies are classically negative, zygous pattern of inheritance.
Type 2 Diabetes Mellitus 301

Wolfram Syndrome MODY type I has decreased HDL cholesterol.


Mutations of HNF4A also occur in the hyperin-
The Wolfram syndrome (#222300 ICD+ sulinemic hypoglycemia in infancy (see Chap.
WOLFRAM SYNDROME 1; WFS1) is caused 12). About two-thirds of the cases of MODY are
by homozygous or compound heterozygous MODY type III. An extremely rare association is
mutation in the gene encoding wolframin (WFS1) MODY type I found with Fanconi renotubular
on chromosome 4p. The syndrome combine fea- syndrome 4 (#616026 FANCONI
tures characterized as DIDMOAD for Diabetes RENOTUBULAR SYNDROME 4 WITH
Insipidus, Diabetes mellitus, Optic Atrophy, and MATURITY-ONSET DIABETES OF THE
Deafness, but only insulin-dependent diabetes YOUNG; FRTS4); remarkably some patients
mellitus and aggressive optic atrophy are essen- present with hypoglycemia rather than hypergly-
tial for the establishment of DIDMOAD. Poor cemia in this association. MODY type II
growth in stature, psychiatric disease, severe neu- (#125851 ICD+MATURITY-ONSET
rological dysfunction, cardiomyopathy, and geni- DIABETES OF THE YOUNG, TYPE 2;
tourinary abnormalities are also described. MODY2) is due to a mutation in the glucokinase
gene. This form presents with mild fasting or
postprandial hyperglycemia which may be man-
Maturity-Onset Diabetes of the Young aged by diet rather than medication. Only 2 %
or Monogenic Diabetes (Table 11.9) will require insulin therapy, possibly during preg-
nancy. MODY type V (#137920 ICD+RENAL
The group of conditions known as maturity-onset CYSTS AND DIABETES SYNDROME;
diabetes of the young (MODY) or monogenetic RCAD) is due to a mutation in the gene encoding
diabetes is caused by at least ten different muta- hepatocyte nuclear factor-1-beta (TCF2, or
tions inherited in an autosomal dominant pattern HNF1B) and combines MODY with cystic dis-
which usually manifest before adulthood or in ease of the kidney and often abnormalities of
the young adult. They are estimated to account male or female genitalia. It may be severe enough
for less than 1–2 % of cases of diabetes although to require kidney transplantation. There are other
it is estimated that 80 % of cases of MODY in seven forms of MODY listed in OMIM where
Great Britain remain undiagnosed. The majority details are available (see Table 11.9).
of cases of MODY are types II and III. MODY MODY should be suspected in patients with
type I (#125850 ICD+MATURITY-ONSET diabetes that have none of the antibodies associ-
DIABETES OF THE YOUNG, TYPE 1; ated with diabetes, in which diabetes at presenta-
MODY1) is caused by a mutation in the gene tion is relatively mild, which appears to be
encoding hepatocyte nuclear factor-4-alpha inherited in an autosomal dominant manner.
(HNF4A), while MODY type 3 (#600496
ICD+MATURITY-ONSET DIABETES OF THE
YOUNG, TYPE 3; MODY3) is caused by a Mitochondrial Disease and Diabetes
mutation in the hepatocyte nuclear factor-1-alpha
gene (142410); some of the manifestations of Maternally inherited diabetes and deafness
both types are similar due to the fact that HNF4- (MIDD) (#520000 ICD+DIABETES AND
alpha regulates the expression of HNF1-alpha. DEAFNESS, MATERNALLY INHERITED;
These patients present with an apparently mild MIDD) can be caused by mutations in several
form of type 2 diabetes, but the condition will different mitochondrial genes: mitochondrial
worsen over years leading to the need for oral tRNA-glutamic acid gene (MTTE), mitochon-
sulfonylurea therapy to avoid the microvascular drial tRNA-leucine 1 gene (MTTL1), or mito-
complications that occur in this condition as well chondrial tRNA-lysine gene (MTTK) which
as in type 1 or type 2 diabetes. MODY type III is impair beta cell function. The diabetes is similar
associated with elevated HDL cholesterol, while to type 2 diabetes in presentation and along with
302 11 Diabetes Mellitus

Table 11.9 Six types of MODY


Condition Gene defect Inheritance Clinical features
MODY 1 #125850 MATURITY- Mutation in the gene Early onset, mild course
ONSET DIABETES OF encoding hepatocyte
THE YOUNG, TYPE I; nuclear factor-4-α
MODY1 (HNF4A; 600281) which
maps to 20q12-q13.1
MODY 2 #125851 MATURITY- Mutation in the gene Early onset, mild course
ONSET DIABETES OF encoding glucokinase common
THE YOUNG, TYPE II; (138079) which maps
MODY2 to 7p13
MODY 3 #600496 MATURITY- Mutation in the hepatocyte Before age 25 years early
ONSET DIABETES OF nuclear factor-1-α gene onset, mild course; insulin
THE YOUNG, TYPE III; (142410), which maps to secretory defect worsens
MODY3 12q34 with age
MODY 4 #606392 MATURITY- Mutation in the IPF1 gene Homozygous infant have
ONSET DIABETES OF (600733) which maps to pancreatic agenesis;
THE YOUNG, TYPE IV; 13q12.2 heterozygous individuals
MODY4 have mild presentation;
rare
MODY 5 #137920 MATURITY- Mutation in the hepatic May have kidney or uterine
ONSET DIABETES OF transcription factor-2 gene defects as well; rare
THE YOUNG TYPE V; (TCF2; 189907), which
MODY5 maps to 17 cen-q21.3.
TYPE V; MODY5
MODY 6/now MATURITY-ONSET Mutation in the Rare, may present with
considered a form of DIABETES OF THE NEUROD1 gene (601724) permanent neonatal
permanent neonatal YOUNG, TYPE VI; MODY6 which maps to 2q31.3 diabetes which may be
diabetes associated with cerebellar
hypoplasia, learning
difficulties, sensorineural
deafness, and visual
impairment

deafness may occur in adulthood. Other features findings of nonketotic hyperosmolar coma are
include findings in the eye (pigmentary retinal severely elevated blood sugar over 600 mg/dL
degeneration and macular pattern dystrophy of and serum osmolality equal to or greater than
the eye retaining normal visual acuity, concentric 350 mOsm/kg; minimal ketosis but the severe
narrowing of visual fields, external ophthalmo- dehydration that results will lead to lactic acido-
plegia), the heart (cardiomyopathy), and central sis which in this case may lead to Kussmaul res-
nervous system (impaired vestibular function and pirations in the absence of ketone bodies. The
dizziness, unsteady gait, seizures, dysarthria). arterial PH should be greater than 7.25 or a
This diagnosis might be suggested by the physi- venous pH greater than 7.3, with a serum
cal findings and a maternal inheritance. bicarbonate greater than 15 mEq/dL for this diag-
nosis. Since the patient has type 2 diabetes and
therefore residual insulin activity even though it
Nonketotic Hyperosmotic Coma is diminished, ketone formation is suppressed
while glucose rises; insulin can suppress ketones
Nonketotic hyperosmolar coma in previous at a lower concentration than required to control
decades was a condition found in adults which hyperglycemia. Any degree of mental disorder up
was also true of type 2 diabetes, but both condi- to coma and seizures may occur. The treatment is
tions are now found in pediatrics. Characteristic rehydration similar to the gradual manner
Summary 303

employed in DKA (above). Serum sodium and appears ever to come closer. Experimental pro-
osmolality will decrease during the rehydration grams have accomplished this goal with small
phase. Insulin at approximately ½ the dose used short-term studies and long-term use may follow.
for DKA may be instituted when blood sugar None of these techniques are yet established or
ceases to decrease at 50 mg/dL/h. approved by the FDA for children, but many are
promising due to the work of many researchers.
Either the biomechanical or biological approach
Summary will likely markedly improve the health of patients
with T1DM in the foreseeable future. The impor-
We have excellent treatment for diabetes but no tant issue is to ensure that they retain good control
cure for diabetes as yet. At present the use of insu- of blood sugar to reduce the development of
lin and high-technology devices are means of comorbidities which will not be reversible even
treating diabetes but not of curing diabetes. Many after we achieve either a cure or improvement in
approaches to finding a cure are under evaluation, treatment.
but none has progressed to established care as yet.
Much research is ongoing into the transplantation
of stem cells to develop islet cells or the transplan- Suggested Readings
tation of actual islet cells into patients with type 1
1. Copeland KC, Silverstein J, Moore KR, Prazar GE,
DM to allow endogenous production and secre-
Raymer T, Shiffman RN, Springer SC, Thaker VV,
tion of insulin in appropriate amounts. Since type Anderson M, Spann SJ, Flinn SK, American Academy
1 diabetes is in most cases immune mediated, of Pediatrics. Management of newly diagnosed type 2
immunomodulatory and anti-inflammatory inter- Diabetes Mellitus (T2DM) in children and adoles-
cents. Pediatrics. 2013;131(2):364–82.
ventions are under evaluation internationally.
2. Sperling MA, Tamborlane WV, Battelino T, Weinzimer
These techniques for diabetes might be offered in SA, Phillip M. Chapter 19—Diabetes mellitus. In:
the future if screening tests suggest a higher risk Sperling MA, editor. Pediatric endocrinology. 4th ed.
for type 1 diabetes or could be utilized after the Philadelphia, PA: Elsevier; 2014. p. 846–900.
3. Sperling MA. Chapter 9—Neonatal diabetes mellitus.
diagnosis during the “honeymoon” period when
In: Sperling MA, editor. Pediatric endocrinology. 4th
beta cell function still exists. Beta cells may be ed. Philadelphia, PA: Elsevier; 2014. p. 277–290.
transplanted or stem cells may be transformed 4. Richardson SJ, Morgan NG, Foulis AK. Pancreatic
into beta cells to be transplanted into an individual pathology in type 1 diabetes mellitus. Endocr Pathol.
2014;25(1):80–92.
with existing type 1 diabetes and immunomodula-
5. Waitzfelder B, Pihoker C, Klingensmith G, Case D,
tion or anti-inflammatory techniques might be Anderson A, Bell RA, Lawrence JM, Mayer-Davis
used to enhance the survival of such beta cells. EJ, Imperatore G, Standiford D, Rodriguez BL,
Alternatively pancreas transplants or partial pan- Dabelea D, Seid M. Adherence to guidelines for
youths with diabetes mellitus. Pediatrics. 2011;
creas transplants might be used in diabetes. An
128(3):531–8.
alternative approach would be the development of 6. Laffel L, Chang N, Grey M, Hale D, Higgins L, Hirst
a “smart” insulin pump that can measure blood K, Izquierdo R, Larkin M, Macha C, Pham T, Wauters
sugar and determine appropriate insulin doses A, Weinstock RS. Metformin monotherapy in youth
with recent onset type 2 diabetes: experience from the
without much intervention from the individual
prerandomization run-in phase of the TODAY study.
with diabetes. We are approaching that level as Pediatr Diabetes. 2012;13(5):369–75.
there is presently a commercial pump which turns 7. Narasimhan S, Weinstock RS. Youth-onset type 2 dia-
the infusion off when it senses a low blood sugar betes mellitus: lessons learned from the TODAY
study. Mayo Clin Proc. 2014;89(6):806–16.
but cannot independently manage diabetes. The
8. Zeitler P, Hirst K, Pyle L, Linder B, Copeland K,
goal of continuous blood glucose monitoring by a Arslanian S, Cuttler L, Nathan DM, Tollefsen S,
miniature device that is linked to the subcutane- Wilfley D, Kaufman F. A clinical trial to maintain gly-
ous insulin infusion pump to form a closed loop cemic control in youth with type 2 diabetes. N Engl J
Med. 2012;366(24):2247–56.
that will function as a replacement pancreas
304 11 Diabetes Mellitus

9. Lee JM, Wu EL, Tarini B, Herman WH, Yoon 18. Achenbach P, Bonifacio E, Koczwara K, Ziegler
E. Diagnosis of diabetes using hemoglobin A1c: A-G. Natural history of type 1 diabetes. Diabetes.
should recommendations in adults be extrapolated to 2005;54 Suppl 2:S25–31.
adolescents? J Pediatr. 2011;158(6):947–52. 19. American Diabetes Association, Lorber D, Anderson J,
e941–3. Arent S, Cox DJ, Frier BM, Greene MA, Griffin Jr JW,
10. Liu E, Lee HS, Aronsson CA, Hagopian WA, Gross G, Hathaway K, Hirsch I, Kohrman DB, Marrero
Koletzko S, Rewers MJ, Eisenbarth GS, Bingley PJ, DG, Songer TJ, Yatvin AL. Diabetes and driving.
Bonifacio E, Simell V, Agardh D, Group TS. Risk of Diabetes Care. 2012;35 Suppl 1:S81–6.
pediatric celiac disease according to HLA haplotype 20. Seaquist ER, Anderson J, Childs B, Cryer P, Dagogo-
and country. N Engl J Med. 2014;371(1):42–9. Jack S, Fish L, Heller SR, Rodriguez H, Rosenzweig
11. Glaser NS, Wootton-Gorges SL, Buonocore MH, J, Vigersky R. Hypoglycemia and diabetes: a report of
Tancredi DJ, Marcin JP, Caltagirone R, Lee Y, a workgroup of the American Diabetes Association
Murphy C, Kuppermann N. Subclinical cerebral and the Endocrine Society. Diabetes Care. 2013;
edema in children with diabetic ketoacidosis random- 36(5):1384–95.
ized to 2 different rehydration protocols. Pediatrics. 21. Dietetics’ AoNa. Pediatric weight management evi-
2013;131(1):e73–80. dence based nutrition practice guidelines. 2007.
12. Glaser NS, Geller DH, Haqq A, Gitelman S, Malloy https://www.andeal.org/topic.cfm?cat=2721
M. Detecting and treating hyperlipidemia in children 22. Dabelea D, Mayer-Davis EJ, Saydah S, Imperatore G,
with type 1 diabetes mellitus: are standard guidelines Linder B, Divers J,… Study S. f. D. i. Y. Prevalence of
applicable to this special population? Pediatr type 1 and type 2 diabetes among children and adoles-
Diabetes. 2011;12(4 Pt 2):442–59. cents from 2001 to 2009. Jama. 2014;311(17):1778–
13. Zeitler P, Haqq A, Rosenbloom A, Glaser N. 1786. doi:10.1001/jama.2014.3201
Hyperglycemic hyperosmolar syndrome in children: 23. Li L, Jick S, Breitenstein S, Michel A. Prevalence of
pathophysiological considerations and suggested Diabetes and Diabetic Nephropathy in a Large U.S.
guidelines for treatment. J Pediatr. 2011;158(1):9–14. Commercially Insured Pediatric Population, 2002–2013.
14.e11–12. Diabetes Care. 2015 Dec 17.
14. Wolfsdorf J, Glaser N, Sperling MA. Diabetic keto- 24. Levitsky LL, Misra M. Epidemiology, presentation,
acidosis in infants, children, and adolescents: a con- and diagnosis of type 1 diabetes mellitus in children
sensus statement from the American Diabetes and adolescents. http://www.uptodate.com/contents/
Association. Diabetes Care. 2006;29(5):1150–9. epidemiology-presentation-and-diagnosis-of-type-
15. Lee YA. Diabetes care for emerging adults: transi- 1-diabetes-mellitus-in-children-and-adolescents.
tion from pediatric to adult diabetes care systems. 25. Levitsky LL, Misra M. Complications and screening in
Ann Pediatr Endocrinol Metab. 2013;18(3): children and adolescents with type 1 diabetes mellitus
106–10. http://www.uptodate.com/contents/complications-
16. Thomas HR, Gitelman SE. Altering the course of and-screening-in-children-and-adolescents-with-type-
type 1 diabetes: an update on prevention and new- 1-diabetes-mellitus.
onset clinical trials. Pediatr Diabetes. 2013; 26. Laffel L, Svoren B. Epidemiology, presentation, and
14(5):311–21. diagnosis of type 2 diabetes mellitus in children and
17. Maassen JA, Janssen GM, t Hart LM. Molecular adolescents. http://www.uptodate.com/contents/
mechanisms of mitochondrial diabetes (MIDD). Ann epidemiology-presentation-and-diagnosis-of-type-
Med. 2005;37(3):213–21. 2-diabetes-mellitus-in-children-and-adolescents.
Hypoglycemia
12

Symptoms of hypoglycemia may be as vague as amino acids (such as alanine derived from muscle),
malaise in a child or feeding difficulty in an infant and lactate, follows. Anaerobic glycolysis (the
or as striking and clinically devastating as grand Embden–Meyerhof pathway) in muscle produces
mal seizures. Conversely, a low blood sugar may pyruvate, which is then metabolized to lactate or
instead be factitious, due to a blood sample stored alanine which are gluconeogenic precursors.
too long in a tube without fluoride before analysis Oxidation of glucose to glycerol-3-phosphate in
or due to the use of outdated test strips for a glu- adipose tissue can also esterify fatty acids in the
cometer. The signs of hypoglycemia revolve synthesis of triglycerides.
around changes in central nervous system func- The muscle mass is smaller in children, and
tion and the autonomic nervous system in the the release of substrates for gluconeogenesis is
absence of glucose. Hypoglycemia should always more limited. Impairment in production of glu-
be confirmed by specific laboratory testing as cose, even with no change in utilization, will
vague feelings of hunger may not be associated reduce serum glucose concentration more quickly
with low blood sugar in spite of common wis- in the child than in the adult. The normal glucose
dom. At the time that glucose decreases, a “criti- production rate is 8 mg per kilogram per minute
cal” blood sample must be obtained (if there is in newborns and infants, 6–8 mg per kilogram
time to briefly and safely delay treatment) or an per minute in young children, and 6–8 mg per
ideal diagnostic opportunity is lost and the diag- kilogram per minute in adolescents as compared
nostic process may become extremely difficult or with 1–2 mg/kg/min in the adult. While the treat-
expensive. ment of disorders of gluconeogenesis will require
an infusion rate of glucose at the normal produc-
tion rate, since treatment simply needs to replace
Normal Carbohydrate Metabolism the glucose that has not been produced, hyperin-
sulinemic states will require a glucose infusion
The maintenance of normal blood sugar depends rate higher than the normal production rate due to
on three basic processes (Fig. 12.1). The intake of the fact that the insulin is actively lowering glu-
carbohydrates from the gastrointestinal (GI) tract cose levels. A large portion of the glucose avail-
initiates the fed state. Glycogenolysis, or the release able in a child is used to provide energy for brain
of glucose from its storage depot, glycogen, is the metabolism. As the brain is much larger in rela-
initial process in the fasted state. The process of tion to body size in the infant than in the adult and
gluconeogenesis, or the production of glucose from consumes relatively more energy, neurologic symp-
precursors such as glycerol (derived from fat), toms and signs of hypoglycemia will be manifest

© Springer International Publishing Switzerland 2016 305


D.M. Styne, Pediatric Endocrinology, DOI 10.1007/978-3-319-18371-8_12
306 12 Hypoglycemia

GH, cortisol Epinephrine

Gluconeogenesis

Glycogen –
+

Liver + Blood
– glucose

Glucagon Insulin
+

Pancreas
+

Glucagon

en
og
Peripheral l yc rgy
G ne
tissues e

Fig. 12.1 Features of carbohydrate metabolism with stimulatory (+) and inhibitory pathways (−) indicated. (Adapted from a
drawing in Endocrinology Nakamoto, JM and Mason, PW. San Juan Capistrano; Quest Diagnostics 2012 with permission)

more readily in the hypoglycemic child. Thus for lates the storage of glucose as glycogen and
many reasons, the child is more susceptible to triglycerides as lipids and promotes the uptake of
hypoglycemia and its symptoms than is the adult. glucose in many other tissues. Most of the uptake
of glucose occurs via facilitated glucose trans-
porters known as GLUT of which there are 14
Glucose Regulatory Factors types, some of which are insulin sensitive and
some of which do not require insulin for their
Concentration of glucose in the serum of normal action. GLUT 4 is insulin sensitive and is found
subjects is kept remarkably constant by gluco- in many tissues including skeletal and cardiac
regulatory factors. Insulin is secreted from the muscle and kidney. Thus insulin decreases blood
beta cells of the pancreatic islets in response to sugar by facilitating the transfer of glucose into
exogenous glucose, which in turn suppresses cells in these organs through GLUT 4 and by
endogenous glucose production by inhibiting decreasing the precursors to gluconeogenesis.
hepatic glycogenolysis and gluconeogenesis (see Glucose is the major source of energy for the
Chap. 11 for discussion of influences on insulin brain. GLUT 1 does not quite require insulin and
secretion). Insulin is a potent suppressor of is found at the blood–brain barrier; transport of
ketone body formation, and as a general rule, if glucose into the brain is directly proportional
ketosis is present in hypoglycemia, abnormally to glucose levels in the circulation. Due to the
increased insulin secretion is not likely the etiol- high glucose requirements of the brain, low
ogy of the hypoglycemia. Further, insulin stimu- plasma glucose concentrations will deprive the
Normal Carbohydrate Metabolism 307

brain of adequate energy. The brain is able to use The Fasting State
ketone bodies as fuel in the absence of adequate In the normal fasting state, insulin secretion is
glucose for at least some of its needs. suppressed so that three processes, mostly resid-
Glucagon is produced and released from the ing in the liver, may support energy metabolism:
alpha cells of the pancreas and quickly but tran- gluconeogenesis (the aerobic production of glu-
siently stimulates hepatic glycogenolysis and cose from the substrate), glycogenolysis (which
gluconeogenesis and also increases hepatic allows the release of glucose from glycogen into
ketone production. Glucagon exerts its effect the circulation), and ketogenesis. Glycogenolysis
through a 7-transmembrane G-protein-coupled is stimulated by rising levels of glucagon and epi-
receptor. Elevated glucagon levels are found in nephrine in the absence of insulin. Glycogenolysis
some stages of uncontrolled diabetes mellitus begins in the fasted state, but glycogen stores are
type 1. In contrast to beta cell destruction which soon exhausted (after about 4 h in infants and
is characteristic of type 1 diabetes, alpha cells after 8 h in older subjects). A disorder of glycoge-
remain as do delta cells which secrete nolysis would cause hypoglycemia early in fast-
somatostatin. ing as will a hyperinsulinemic condition. The
Epinephrine from the adrenal medulla can next process leading to hypoglycemia if disor-
rapidly stimulate hepatic glycogenolysis and dered is gluconeogenesis which utilizes amino
gluconeogenesis acting through adrenergic G acids derived from the breakdown of muscle pro-
protein membrane receptors, indirectly affecting tein and lactate from blood cell glycolysis (the
insulin and glucagon as well. process which converts glucose to lactate). To
Both the cortisol and growth hormone (GH) preserve muscle protein, hepatic fatty acid oxida-
are considered to be counterregulatory hormones tion next provides the energy for gluconeogene-
and exert anti-insulin effects by limiting glucose sis. Lipolysis breaks down lipids into triglycerides
utilization and increasing glucose production. which are metabolized into glycerol and free
GH, however, has an additional early, transient fatty acids (FFAs). Lipolysis is stimulated by
effect of decreasing serum glucose through the growth hormone, cortisol, and epinephrine. Fatty
production and action of the insulin-like growth acid oxidation becomes prominent 8–12 h after
factors, although the ultimate effect of GH is to fasting in the neonate but only after about 16–24 h
elevate glucose concentrations (Table 12.1). in older children.
The liver is also the site of formation of the
The Fed State ketone bodies acetoacetate (which is converted
After a meal, in the postprandial state, exogenous to acetone) and β-hydroxybutyrate from FFAs
glucose is used by, and stored in, tissues, whereas through mitochondrial fatty acid oxidation dur-
the mechanism of glucose production, gluconeo- ing the breakdown of lipids. The ketone bodies
genesis, is suppressed in normal conditions. can pass the blood–brain barrier and provide
Thus, insulin secretion increases and glucagon energy for brain function, albeit limited energy,
decreases with glucose intake in the fed state. and can be used for energy in the skeletal and
Insulin is responsible for the storage of glucose cardiac muscle. The production of ketone
through the production of glycogen mainly in the bodies by fatty acid oxidation may be viewed as
liver and muscle, the transport of amino acids, an adaptive mechanism that spares muscles from
and incorporation of amino acids into protein in breakdown and the release of amino acids that
muscles and lipids into triglycerides in the adi- would otherwise be used for gluconeogenesis;
pose organ. Insulin suppresses the production of likewise, the process preserves glucose to sup-
ketone bodies; ketone bodies are very low or port brain metabolism and to allow glucose-
unmeasurable in blood in the fed state. In fact, dependent metabolism in the red cells. In a
insulin is more efficient at suppressing ketone similar manner, the energy needs of some organs
formation than it is in decreasing blood sugar. can shift to conserve glucose as the liver, adipose
Table 12.1 Causes of hypoglycemia
Free
Condition Insulin Ketones GH Cortisol Lactate fatty acids GIR Other features
Transient hypoglycemia Low NI or high Appropriate Appropriate NI or high NI or high 6–10 mg/kg/min
of the newborn for BS for BS
Persistent High for glucose Low Appropriate Appropriate Low Low >12–15 mg/kg/min Some defects
hyperinsulinemic value for BS for BS associated with
hypoglycemia of infancy elevated ammonia,
IGF BP1 is low
Beckwith-Wiedemann High for glucose Low Appropriate Appropriate Low Low >12–15 mg/kg/min Organomegaly,
syndrome value for BS for BS elevated IGF-2,
IGF BP1 is low
Beta cell adenoma High for glucose Low Appropriate Appropriate Low Low >12–15 mg/kg/min IGF BP1 is low
value for BS for BS
GH deficiency Nl High Stimulated peak, Nl (if GH High High 6–10 mg/kg/min May have
<10 ng/mL deficiency is findings of SOD
isolated)
ACTH or cortisol Nl High Nl Stimulated peak, High High 6–10 mg/kg/min May have
deficiency <15–18 μcg/mL findings of SOD
Galactosemia Low High Nl Nl High High 6–10 mg/kg/min High galactose
Acyl-CoA Nl Low Nl Nl High High 6–10 mg/kg/min Low carnitine
dehydrogenase,
medium-chain deficiency
Glycogen storage Low High Nl Nl High High 6–10 mg/kg/min Hepatomegally in
disease types 1, 3, 6, 9, 11, 0 type 1, 3, 6, 9, 0
Ketotic hypoglycemia Low High Nl Nl High High 6–10 mg/kg/min Low alanine
(could be GH or cortisol
deficiency)
Hereditary fructose Low High Nl Nl High High 6–10 mg/kg/min Immediately
intolerance precipitated by
fructose intake
Fructose 1-6 Low High Nl Nl High High 6–10 mg/kg/min Not immediately
diphosphatase deficiency precipitated by
fructose intake
but by fasting
Munchausen by proxy High for glucose Low Nl Nl Low Low >12–15 mg/kg/min
with exogenous insulin but C peptide is low until elimination of
administration exogenous insulin
GH growth hormone, Nl normal, ACTH adrenocorticotropic hormone, GIR glucose infusion rate, SOD septo-optic dysplasia
Adapted from a chart in Endocrinology Nakamoto, JM and Mason, PW. San Juan Capistrano; Quest Diagnostics 2012 and Selected Readings
Hypoglycemia 309

tissue, and muscle can use the β-oxidation of An infant showing signs of hypoglycemia
fatty acids. should have a laboratory glucose determination,
In the fasted state, the endocrine milieu main- a critical blood sample drawn (see below), and
tains blood sugar concentration at about 55 mg/ glucose provided. If there is a medical history or
dL: if the blood sugar falls below 55 mg/dL, physical findings suggesting a hypoglycemia
endocrine factors attempt to raise blood sugar to syndrome, this will assist in the diagnosis. The
compensate. A “critical sample” will indicate AGA baby who has prolonged hypoglycemia
which hormone concentrations rise appropriately after 24 h of age is likely to have a disease as eti-
to increase blood sugar or which are inadequate ology of hypoglycemia.
to do so (e.g., GH or ACTH deficiency) or if
hyperinsulinism is driving blood sugar down.
The critical sample is described below. Hypoglycemia (Table 12.1)

Symptoms of hypoglycemia are due either to


Transition of the Fetus effects of adrenergic and cholinergic agents
to Neonatal Life (causing sweating, tremulousness, hunger, weak-
ness, and tachycardia) or to central nervous sys-
The fetus is dependent upon maternal supplies of tem (CNS) manifestations (such as headaches or
glucose, and serum levels of glucose in the fetus reduced mentation, which may extend on a con-
vary according to the mother’s levels. Thus a tinuum from drowsiness to coma and seizures).
mother with diabetes who experiences hypogly- Even if severe and recurrent hypoglycemia is
cemia has a fetus who likewise experiences ultimately controlled, the patient may still be left
hypoglycemia, and the opposite occurs when the with permanent neurologic impairment such as
mother’s blood sugar is too high. The eight developmental delay, memory loss, and seizure
GLUTs found in the placenta are mostly insulin disorders that manifest even when the blood
independent through most of pregnancy, and thus sugar is normal. Patients taking adrenergic block-
the transport of glucose for mother and child ing agents may not show the initial symptoms of
depends upon the concentration gradient rather hypoglycemia in spite of having low blood sugar
than upon insulin levels. When the child is sepa- concentrations, but still have the potential for
rated from the maternal glucose supply at birth, neurologic damage. Symptoms of hypoglycemia
mobilization of glucose from stored sites must usually begin only when blood sugar decreases
begin immediately and gluconeogenesis must below 55–68 mg/dL, so a value of 55 mg/dl
commence. During this transitory period, almost should trigger an evaluation. Repeated hypogly-
1/3 of appropriate-for-gestational-age newborn cemia decreases the release of epinephrine from
will experience glucose levels less than 50 mg/dL the adrenal medulla leading to hypoglycemia-
during the first day of postnatal life in the absence associated autonomic failure (HAAF) and hypo-
of disease or symptoms; there is rarely a need to glycemic unawareness since the adrenergic
monitor glucose levels in a normal-term newborn response is one of the features that make a hypo-
who had a normal delivery from a healthy mother glycemic individual realize that he or she is
according to most authorities. More significant hypoglycemic. HAAF can occur in repeated
hypoglycemia in the first 24 h may occur in hypoglycemia or in the hypoglycemia that occurs
small-for-gestational-age infants, premature with the treatment of diabetes mellitus as
babies, babies suffering from neonatal stress or described in Chap. 11. The endocrine response to
asphyxia, and of course infants of diabetic moth- hypoglycemia might occur at lower glucose level
ers or infants with increased insulin secretion. It than 55 mg/dL after repeated episodes of hypo-
is sometimes difficult to determine who is having glycemia which further complicates the diagno-
transitional mild hypoglycemia in the first 24 h sis of the etiology of hyperglycemia since the
versus those with a condition requiring treatment. secretion of growth hormone and cortisol as
310 12 Hypoglycemia

counterregulatory hormones will not be triggered Thus symptoms of hypoglycemia in the newborn
until blood sugar drops below the usual threshold range from grand mal or localized seizures to irri-
of hypoglycemia of 55 mg/dL. tability, hypotonia, lethargy, and difficulty in
The classic method of establishing hypoglyce- feeding, to other rather nonspecific findings, and
mia invokes Whipple’s triad. Thus the symptoms to no findings at all because of the immaturity of
or signs of hypoglycemia must occur with a con- the CNS. A high index of suspicion for hypogly-
firmed low blood sugar measurement and must cemia must be maintained especially with certain
be resolved when the glucose rises. Whipple’s clinical conditions known to be associated more
triad was developed for adults but appears appro- frequently with hypoglycemia.
priate for children and teenagers but not neces- The neonate has less tolerance to fasting and
sarily neonates in whom symptoms are more stress than does an older individual because of
subtle. While the threshold 55 mg/dL is appropri- limited glycogen or gluconeogenic substrate
ate for the diagnosis of hypoglycemia, treatment availability, and hypoglycemia develops faster in
should aim to maintain a blood sugar greater than the newborn. Disorders causing hypoglycemia in
70 mg/dL. the newborn can be found on a continuum from
Various etiologies of hypoglycemia tend to decreased ability to stabilize metabolically in the
emerge in a certain chronological order in the postnatal period, continuing to hypoglycemia as
postprandial state after fasting begins, and the a secondary finding associated with other dis-
timing of hypoglycemia provides an important eases, and ending with disorders in which severe,
clue to the diagnosis. For example, hypoglycemia unremitting hypoglycemia is the immediate and
due to fructose intolerance manifests just after primary problem.
ingesting fructose, and leucine-sensitive hyperin- Even a normal newborn who has fasted for
sulinism likewise occurs after the ingestion of 6 h has a 10 % chance of having a blood sugar
leucine. Glycogen storage disease (GSD), in less than 30 mg/dL and a 30 % chance of devel-
which there is difficulty in glycogenolysis, mani- oping a blood sugar less than 50 mg/dL, so tran-
fests in a few hours after eating. After a period of sient hypoglycemia is common. Availability of
fasting, other forms of hyperinsulinism or a dis- ketone bodies as alternative fuel for the CNS
order of glycogenolysis will be manifest; next appears to mitigate long-term effects of this
will appear disorders of gluconeogenesis includ- transient hypoglycemia. A neonate who is the
ing growth hormone and cortisol deficiency or product of a problem pregnancy or delivery is an
fructose-1,6-bisphosphatase deficiency; lastly, even more likely candidate for transient hypo-
disorders of fatty acid oxidation (acyl-CoA dehy- glycemia. Thus infants who are premature or
drogenase deficiencies) will emerge during a pro- small for gestational age, or who experienced
longed fast. trauma, asphyxia, or cold exposure at birth,
should be monitored prospectively for the devel-
opment of hypoglycemia. Diseases that have
Hypoglycemia in the Newborn other significant manifestations may have hypo-
glycemia included in the constellation; frequent
The definition of hypoglycemia in the newborn examples include sepsis, postexchange transfu-
has been controversial, but a plasma glucose less sion or erythroblastosis fetalis, congenital heart
than 50 mg/dL or, because whole-blood glucose disease, or congenital defects of the CNS or
is 15 % lower than serum or plasma glucose, a elsewhere. The condition of the mother before
blood glucose less than 43–45 mg/dL may be delivery may affect the neonate cell; for exam-
used for the diagnosis of hypoglycemia in the ple, infants of mothers with toxemia, with nar-
symptomatic neonate in the first 24 h after birth. cotic addiction, or who are taking oral
Unlike hypoglycemia found in older children or hypoglycemic agents or β-adrenergic blockers
teenagers, symptoms of hypoglycemia in the are likely candidates for hypoglycemia as are
newborn are more generalized and may be subtle. infants of diabetic mothers.
Hypoglycemia 311

Hyperinsulinemic Hypoglycemia Mothers with permanent or gestational diabetes


in the Newborn mellitus (GDM) who have not maintained eugly-
A child who is a term product of a normal deliv- cemia may have infants who experience intrauter-
ery without known complication, fed by 6 h after ine hyperglycemia, related to maternal blood sugar
birth, should not have persistent hypoglycemia. concentrations, and who develop beta cell hyper-
However in the presence of excessive insulin plasia in the islets of Langerhans because of this
secretion, glucose falls as it is utilized in the met- stimulation by ambient glucose concentrations;
abolic processes described above promoted by after birth, when removed from the maternal glu-
insulin. Infants of diabetic mothers have a risk cose supply, the increased insulin secretion of the
for hyperinsulinemic hypoglycemia, but it will islets will cause minimal to severe hypoglycemia
be of a transient nature during the first week after for hours to a few days after delivery. The IDM,
birth. Newborns who are not infants of diabetic unless having intrauterine growth restriction
mothers with persistent hypoglycemia after age (IUGR) due to even more severe maternal diabe-
2 days and who require more than 10 mg/kg/min tes, has a characteristic appearance of increased
of intravenous glucose to maintain a normal size for gestational age (they are longer as well as
blood sugar most likely have the hypoglycemia heavier), with extra subcutaneous tissue, a pletho-
because of a disorder causing hyperinsulinism of ric complexion, and a rather lethargic level of
the newborn and will have a more difficult course activity. The hypoglycemia is not solely due to
than those with the transient problems mentioned. elevated insulin as they also have decreased gluca-
Hyperinsulinism may cause devastating hypogly- gon and catecholamine production.
cemia in the newborn or older child. Diagnosis is These infants will require a glucose infusion
suspected if any insulin secretion is detected dur- rate in the range of 10–12 mg per kilogram per
ing hypoglycemia, a state in which normally no minute and sometimes as high as 15 mg per kilo-
insulin should be measurable; even low levels of gram per minute. This condition is less frequent if
insulin secretion while the blood sugar is low may the mother has excellent blood sugar control dur-
suggest hyperinsulinism. Hyperinsulinism is ing pregnancy. Maternal medications including
especially likely to cause brain damage, as an sulfonylureas, terbutaline, and propranolol can
absence of glucose as well as lactate and ketone cross the placenta and cause neonatal hypoglyce-
bodies and found, so that all metabolic fuels for mia in the newborn. Similarly, infants of mothers
the brain are unavailable. Absence and suppres- who are not diabetic but were receiving a large
sion of serum ketone bodies and FFAs are diag- amount of intravenous glucose will have increased
nostic clues to hyperinsulinism. fetal insulin secretion; because the glucose will
Nesidioblastosis was a term classically used to abruptly disappear at the time of placental separa-
describe newborns with hyperinsulinemia which tion, the child is also at risk for transient hyper-
was also called islet cell hyperplasia, islet cell insulinemic hypoglycemia. In all of these cases,
dysplasia, and islet cell dysmaturity; it represents hypoglycemia should have resolved within a few
the histologic finding of beta cells in the pancre- days or there will be another diagnosis indicated.
atic ductal tissue. Nesidioblastosis was defined as Persistent hyperinsulinemic hypoglycemia in
the development of beta cell islets from pancre- infancy may occur due to mutations of a variety
atic duct tissue, but more recent histologic stud- of genes causing seven different forms of the
ies suggest that this appearance is a normal condition. All forms are due to defective feed-
variant; some of the patients previously diag- back inhibition of glucose upon insulin secretion
nosed as having nesidioblastosis may have had due to mutations in the beta cell; we will consider
microscopic adenomas that had been missed or a five forms here but more may be found in refer-
functional defect that could not be demonstrated ences at the end of the chapter or in OMIM. The
histologically but can now be demonstrated by etiology of these conditions involves mutations
molecular probes as noted below. in the process in which the beta cell senses
312 12 Hypoglycemia

glucose and releases insulin. In the normal situation, to suppress insulin secretion. There is heteroge-
glucose is taken up by the beta cell by the action neity in these conditions so that some require
of the GLUT2 transporters and phosphorylated pancreatectomy and some respond to diazoxide
by the enzyme glucokinase. Glucose is oxidized, with the same gene defect differing in therapeutic
and the plasma membrane ATP-dependent potas- requirements if the inheritance is autosomal
sium (KATP) channel is closed by increased recessive or dominant.
ATP/ADP ratio which leads to the voltage- Leucine will also raise the ATP/ADP ratio,
dependent calcium channel to now allow the closing the channel and leading to insulin secre-
influx of calcium into the cell, ultimately leading tion, and mutations may cause leucine-sensitive
to insulin secretion. The KATP channel consists hypoglycemia.
of a K1-selective pore-forming subunit (Kir6.2) The first described form of hyperinsulinemic
and a regulatory subunit (SUR-1) and mutations hypoglycemia of the newborn (#256450 ICD+
in either of which occur in persistent hyperinsu- HYPERINSULINEMIC HYPOGLYCEMIA,
linemic hypoglycemia in infancy (Fig. 12.2). The FAMILIAL, 1; HHF1) may be inherited in an
KATP channel is closed by the class of oral dia- autosomal recessive or an autosomal dominant
betes medications, sulfonylureas, which stimu- pattern and results from a mutation in the ATP-
late insulin secretion and opened by diazoxide binding cassette, subfamily C, member 8 gene
which is used in certain conditions listed below (ABCC8) which affects the function of the KATP

ATP-sensitive K+ channel
Ca2+
K+ Voltage
dependent
Ca2+ channel
SUR1
Depolarization
Mitochondria Ca2+
6. R
KU
2

ATP/ADP↑ cAMP
Nucleus

Islet
Pyruvate transcription
factors
Glucose-6-phosphate
Glucokinase
Glucose
Secretory
granules Insulin

Glucose

GLUT2

Fig. 12.2 The beta cell and its components. The ATP- duce glucose-6-phosphate. Glucose-6-phosphate can be
sensitive K+ channel (KATP) is the site of action of sulfo- metabolized during glycolysis leading to increased ATP
nylureas which close the channel to cause insulin secretion which enhances insulin secretion through the ATP-
and of diazoxide which opens the channel to suppress sensitive K+ channel. Thus glucokinase acts as the
insulin secretion. Mutations in this area are responsible glucose sensor of the beta cell and plays a major role in
for some forms of hyperinsulinemic hypoglycemia of the determining how much insulin is released in response to
newborn. The GLUT 2 transporter allows glucose to enter glucose levels in the normal condition. Mutations in
the beta cell where it interacts with glucokinase to pro- glucokinase can lead to hypoglycemia or hyperglycemia
Hypoglycemia 313

channel. The patients are large due to the stimula- in the fasting or fed state, and surgery may not be
tion of the IGF-1 receptors by excessive insulin necessary. Leucine intake must be minimized.
during fetal life and later in life may develop Leucine can also precipitate hyperinsulinemic
insulin deficiency and diabetes rather than con- hypoglycemia in another condition in an autosomal
tinuing hyperinsulinism. A single mutation may dominant pattern (240800 HYPOGLYCEMIA,
exist on the paternal chromosome, with loss of LEUCINE-INDUCED) due to mutation in
the normal maternal allele leading to homozy- the ATP-binding cassette, subfamily C, member
gosity for a recessive SUR1 mutation, leading to 8 gene (ABCC8) affecting the SUR-1 subunit.
focal pancreatic disease. This condition most Hyperinsulinemic hypoglycemia type 7
often does not respond to diazoxide but may be (#610021 HYPERINSULINEMIC
amenable to surgical correction without the need HYPOGLYCEMIA, FAMILIAL, 7; HHF7)
for total pancreatectomy. However, there is a manifests as hypoglycemia after exercise is due
dominant subtype with normal birth weight to elevated pyruvate levels. This condition is due
which does respond to diazoxide. The type 2 to mutations in the solute carrier family 16, mem-
form (#601820 ICD+ HYPERINSULINEMIC ber 1 gene (SLC16A1). The clinical findings are
HYPOGLYCEMIA, FAMILIAL, 2; HHF2) is variable but some patients have been treated suc-
due to a mutation in the Kir6.2 subunit of the cessfully with partial or total pancreatectomy and
potassium inwardly rectifying channel (KATP others with diazoxide or by following a sedentary
channel), subfamily J, member 11 gene (KCNJ11) lifestyle.
which is inherited in an autosomal recessive pat- The glucose infusion rate required for
tern, and birth weight is usually high. Both homo- successful management of hyperinsulinemic
zygous dominant HHF type I and HHF type II are hypoglycemia most often will be high, e.g., 10 or
unlikely to respond to diazoxide therapy since 12 mg per kilogram per minute, which must be
the site of the defect is where diazoxide exerts its maintained until success with medical or surgical
effect but heterozygous recessive forms may therapy is accomplished. Up to 30 % of patients
respond to diazoxide. with hyperinsulinism are reported to have devel-
Hyperinsulinemic hypoglycemia type 3 is auto- opmental delay probably related to delay in diag-
somal dominant (#602485 HYPERINSU- nosis and therapy. Patients with autosomal
LINEMIC HYPOGLYCEMIA, FAMILIAL, 3; recessive abnormalities in the KATP channel are
HHF3) and is due to a mutation of the glucokinase not likely to respond to diazoxide since the KATP
gene. HHF3 may respond to oral diazoxide therapy. channel is the location of diazoxide action.
Glutamate dehydrogenase hyperinsulinism or However, there are dominantly inherited muta-
hyperinsulinemic hypoglycemia type 6 (#606762 tions in the KATP channel (mutations of ABCC8
HYPERINSULINEMIC HYPOGLYCEMIA, and KCNJ11) which may respond to diazoxide at
FAMILIAL, 6 at 10q23.3; HHF6) manifests ele- a dose of 5–15 mg/kg/day divided into two or
vated ammonia levels about 2–5 times the nor- possibly one dose as indicated above.
mal. This condition may be discovered later in Diazoxide was developed to lower blood pres-
life rather than infancy. The condition is due to sure and also causes fluid retention, so a thiazide
mutations in the glutamate dehydrogenase gene diuretic is sometimes necessary to offset the fluid
(GLUD1) which is involved in ammonia metabo- retention. Likewise, over long use, diazoxide
lism. Leucine activates the enzyme and causes increases hair growth to a quite noticeable degree,
insulin secretion normally, but in this condition, particularly on the face. If diazoxide is ineffec-
inhibition of this process is defective, leading to tive, octreotide may be tried in a dose of 5–20 mg/
excessive insulin secretion. This condition is kg/day given subcutaneously in divided doses
inherited in an autosomal dominant pattern con- 3–4 times per day or intravenously by a continuous
dition (although 80 % are new mutations and infusion by pump. Long-acting forms of octreo-
appear to be sporadic). Treatment with diazoxide tide such as used in acromegaly are sometimes
is often successful in controlling hypoglycemia effective in older children. Octreotide treatment
314 12 Hypoglycemia

carries the risk of necrotizing enterocolitis and mal at birth. Adrenal hypoplasia, dysplasia, or
development of gall bladder sludge and stones; atrophy will not necessarily have diagnostic
ursodiol may be used to control the latter. physical findings but will lead to hypoglycemia
Some of the hyperinsulinemic hypoglycemic (see Chap. 3).
syndromes present with focal lesions in the pan- Fructose-1,6-bisphosphatase deficiency
creas, while others have diffuse pancreatic (#229700 ICD+ FRUCTOSE-1,6-BISPHOS-
involvement. If medication is unsuccessful and PHATASE DEFICIENCY at 9q22.3) is the result
surgery is indicated, it is possible to separate of a defect in fructose-1-phosphate aldolase,
patients with local lesions that may be amenable which utilizes lactate, alanine, glycerol, and oral
to partial pancreatectomy rather than diffuse fructose for gluconeogenesis. Along with hypo-
lesions requiring total pancreatectomy by the glycemia, the newborn infant may have hyper-
use of imaging techniques. While standard ventilation, apnea, seizures and coma,
CT scans or MRI imaging will not usually reveal hepatomegaly, elevated uric acid, and ketosis or
the focal lesions, positron emission tomogra- metabolic acidosis. The liver is enlarged as in
phy (PET) scanning using fluorine-18 L-3, GSD type 1, in this condition because of hepatic
4-dihydroxyphenylalanine (18F-fluoro-L-DOPA) steatosis, and differential diagnosis between the
tracer takes advantage of the fact that the lesions two conditions is through the fed glucagon stimu-
take up L-dopa to demonstrate the local lesions. lation test (administration of glucagon after
Hypoglycemia may be caused by the hyper- meal). Signs and symptoms appear with fasting
insulinism characteristic of the Beckwith– or illness rather than directly after fructose inges-
Wiedemann syndrome (#130650 BECKWITH- tion. A newborn with hypoglycemia after fasting
WIEDEMANN SYNDROME; BWS at 11p15.5), with lactic acidosis and inadequate rise of blood
whose features include exomphalos, macroglos- sugar to glucagon administration is the classic
sia, and gigantism or in the infant, giant syndrome presentation. Determination of a mutation in the
(large body size and microcephaly but no macro- fructose-1,6-bisphosphatase gene (FBBP 1) is
glossia or exomphalos). diagnostic; minimizing fasting and decreasing
Beta cell adenomas are extremely rare in fructose intake as well as avoiding sorbitol is the
infants and newborns and, although more com- management. The condition is autosomal
mon in later childhood, are still rarer than in recessive.
adults. Hyperinsulinism of this type may be a Hereditary fructose intolerance (#229600
part of multiple endocrine neoplasia type 1 but is ICD+ FRUCTOSE INTOLERANCE,
usually found well after the second decade (see HEREDITARY) presents with hypoglycemia,
Chap. 10). abdominal pain, vomiting, and diarrhea after
exposure to fructose which may first occur after
Defects in Gluconeogenesis weaning. The child may demonstrate aversion to
in the Newborn sweet tastes. Laboratory determination reveals
Deficiencies of GH and/or adrenocorticotropic hypoglycemia, and with continued exposure to
hormone (ACTH) may cause neonatal hypogly- fructose, the concentrations of uric acid, lactic
cemia. In boys, the finding of microphallus (penis acid, and magnesium rise, while phosphorus,
less than 2 cm in length due to gonadotropin and/ potassium, and bicarbonate fall. If the fructose
or GH deficiency) associated with hypoglycemia dose increases or exposure continues shock, hep-
should strongly suggest this diagnosis; visual atomegaly, hepatic steatosis, and ultimately cir-
impairment and optic hypoplasia also may be rhosis and liver failure occur and later kidney
found in septo-optic dysplasia (see Chap. 3). failure will develop. This is a severe condition
Virilizing congenital adrenal hyperplasia will with a potentially fatal outcome. Intravenous glu-
often present with hypoglycemia and cortisol cose is the appropriate early treatment, and
deficiency but will be indicated by ambiguous avoidance of all fructose and sorbitol in the diet
genitalia in girls although boys will appear nor- is required. It is not recommended that a child
Hypoglycemia 315

undergoes fructose challenges as the danger is too develops in the newborn or infant period, but
great. The disorder is caused by a mutation in the sometimes patients are well until several years of
aldolase B, fructose-bisphosphate gene (ALDOB) age. Hypoglycemia occurs after prolonged fasts
and inheritance is autosomal recessive. that may accompany intercurrent illnesses.
Metabolic abnormalities of branched-chain Hyperammonemia, hyperuricemia, acidosis, and
amino acids, such as maple syrup urine disease elevations of liver enzymes are rarely seen in this
(*248600 MAPLE SYRUP URINE DISEASE, condition. This may present in a similar manner to
TYPE IA at 19q13.1-q13.2), are found in an auto- Reye syndrome, with disordered mentation and
somal recessive pattern and characterized by ele- absent ketone bodies during hypoglycemia.
vated branched-chain amino acids (leucine, Plasma carnitine is low, and an increased ratio of
isoleucine, or valine) in serum and urine. In esterified carnitine to free carnitine is found.
affected infants, developmental delay, coma, sei- Medium-chain dicarboxylic acid should be mea-
zures, vomiting, lethargy, elevated serum ammo- sured in the urine to make the specific diagnosis.
nia, and metabolic acidosis in the presence of Diagnosis can be accomplished through determi-
ketotic hypoglycemia will be found. The urine has nation of the types of acyl-carnitine present by
the odor of maple syrup (as suggested by the name mass spectroscopy, a technique now becoming
of the disorder). Five different forms and degrees more frequently used in newborn screening pro-
of severity are found, with the basic defect in all grams. Life-threatening emergencies may be
being mutations in the branched-chain α-keto acid avoided by early diagnosis. The treatment usually
dehydrogenase, E1α subunit gene. Other forms of is elimination of fasting for more than a few hours,
maple syrup urine disease are termed type 1B 6–8 h in infants and 10–12 h for older children.
(*248611) and type 2 (*448610). Primary carnitine deficiency (#212140 ICD+
Galactosemia (#230400 GALACTOSEMIA at CARNITINE DEFICIENCY, SYSTEMIC
9p13) is one of the diseases for which generalized PRIMARY; CDSP) is due to a loss-of-function
screening is available at birth. Only at the late mutation in the solute carrier family 22 (organic
stages of the disease in which liver failure devel- cation transporter), member 5 gene (SLC22A5)
ops is hypoglycemia apparent. The diagnosis will and is inherited in an autosomal recessive pattern.
usually rest on findings other than low blood The form presenting in infancy leads to acute
sugar such as cataracts, hepatomegaly, decreased hypoglycemic episodes without ketosis, while if
liver function, vomiting, diarrhea, hemolytic ane- the onset is in childhood after 1 year, there is pro-
mia, and ultimately ovarian failure in females. gressive cardiomyopathy and muscle weakness.
Disorders of organic acid or fatty acid oxida- There are numerous other features if the condition
tion represent a more common type of disorder is not treated involving the heart (cardiomegaly,
(with a prevalence of one in 9,000–15,000), lead- hypertrophic cardiomyopathy, congestive heart
ing to fasting hypoglycemia of various degrees failure), the liver (hepatomegaly, steatosis), and
extending to a life-threatening condition. These the musculoskeletal system (muscle weakness)
disorders limit the production of ketone bodies which include the following: hypotonia; muscle
during fasting, so they mimic the hypoglycemic biopsy shows lipid deposition; lethargy associated
hyperinsulinemic disorders in this regard and with hypoglycemia; encephalopathy associated
deprive the brain of both glucose and ketone bod- with hypoglycemia; coma associated with hypo-
ies for energy. The conditions are generally the glycemia; Reye syndrome; delay in gross motor
result of defects in the acyl-CoA system, with development due to weakness; fatty infiltration of
short-, medium-, or long-chain fatty acids heart muscle; abnormal liver enzymes; increased
involved in the various classifications. The most urinary carnitine; decreased carnitine in serum,
common type is medium-chain acyl-CoA dehy- muscle, heart, and liver; impaired carnitine uptake
drogenase (MCAD) mutations (#201450 ACYL- in muscle, heart, kidney, fibroblasts, and lympho-
CoA DEHYDROGENASE, MEDIUM-CHAIN, cytes; hyperammonemia; no dicarboxylic acid-
DEFICIENCY OF at 1p31). Hypoglycemia uria; and no ketosis.
316 12 Hypoglycemia

Hypoglycemia in Older Children may lead to a sharp increase in serum glucose,


causing release of insulin that subsequently
A diagnosis of hypoglycemia in older children causes hypoglycemia, but patients will have a
rests upon a higher threshold than during the first history of GI surgery as an indication of this con-
24 h after birth. Hypoglycemia may be diagnosed dition; this occurs more often if the G-tube infu-
at a plasma blood sugar lower than 55 mg/dL sion is administered in boluses rather than
(blood sugar less than 47 mg/dL). However, for continuously. A dumping syndrome constellation
the purposes of treatment, the goal should be may be found with the increasing use of gastric
plasma sugar greater than 70 mg/dL (blood sugar bypass surgery in adolescents with extreme obe-
greater than 60 mg/dL). sity. Dietary measures may be used such as corn-
Some of the same conditions noted under starch bars which are used in children with
hypoglycemia of the newborn may be diagnosed diabetes to slow digestion and maintain blood
as causes of hypoglycemia in later life. Some of sugar. These conditions may be successfully
the conditions described below can present in treated with somatostatin agonists (e.g., octreo-
newborn in a relatively subtle fashion so that the tide) to suppress insulin secretion or by an alpha
diagnosis is initially missed and only diagnosed glucosidase inhibitor (acarbose).
later in life as the severity of the hypoglycemia is Delayed insulin secretion is reported in rare
noted. The time period between feedings length- patients who ultimately develop type 1 diabetes
ens with age, leading to more opportunities for mellitus. In contrast, “reactive hypoglycemia” is
fasting hypoglycemia to present, and the onset of all too often diagnosed in children who have
common illnesses that decrease feeding fre- unusual behavior or sensations attributed to low
quency likewise can precipitate a hypoglycemic serum glucose but without confirmatory mea-
episode. There is however a change in the preva- sured glucose values; sometimes parents pressure
lence of the disorders with advancing age. For the clinician to make the diagnosis because they
example, hyperinsulinism of infancy occurs at a think that they have hypoglycemia themselves
younger age, while GSD, other than type 1 GSD, (without laboratory evidence) or because they
becomes more frequently diagnosed at an older have read the latest popular book on the subject.
age, and beta cell adenomas replace most types Insulin resistance can occur because of con-
of familial hyperinsulinemic hypoglycemia in the genital defects or because of acquired conditions
differential diagnosis as children become older. including obesity and polycystic ovarian syn-
In all age groups, obtaining the critical sample is drome. While the body is generally resistant to
of great importance in the differential diagnosis. the extremely high levels of insulin which occur
in insulin resistance, continued secretion of insu-
Hyperinsulinism lin or decreased clearance of insulin from the cir-
Hyperinsulinemic hypoglycemia of infancy is culation as blood sugar is falling leads to
described above. Some patients will present later hypoglycemia in some patients. Diagnosis will
in life although they have a congenital defect. be made with a prolonged, 5-h, glucose tolerance
Some will actually have experienced hypoglyce- test with measurement of glucose and insulin
mia to an extent which causes brain damage with each blood sample; accurate results require
before the diagnosis is made. that the child has been on a high carbohydrate
Hyperinsulinism will be more likely the result diet for at least 3 days before the test. Symptoms
of a β-cell adenoma after the newborn period the child may feel are observed and recorded to
than of a congenital condition, especially after see if these symptoms chronologically relate to
age 3 years. This rare but serious condition may any low glucose values detected. If the symptoms
be difficult to diagnose by imaging techniques. It can be reversed with glucose administration, the
may be found as one of the manifestations of condition is more definite. This is the application
MEN type I (Wermer syndrome) (see Chap. 10). of Whipple’s triad.
Rapid gastric emptying resulting from the Congenital hyperinsulinism due to mutations
dumping syndrome after Nissen fundoplication in the insulin receptor (INSR) can present with a
Hypoglycemia 317

variety of physical features. The child is charac- Hypoglycemia with Ketosis


teristically small for gestational age and has Ketotic hypoglycemia classically is seen in a
acanthosis nigricans as a manifestation of insulin thin (often male) child of age 18 months to
resistance. The Donohue syndrome is one exam- 5 years, who has had a longer-than-average
ple of congenital hyperinsulinism (#246200 overnight fast (sometimes this is called the
ICD+ DONOHUE SYNDROME) leading to Saturday night–Sunday morning syndrome
postprandial or fasting hypoglycemia. The because of the purported late return of parents at
patients present with a phenotype of large ears night, causing a delay in breakfast the following
and “elfin” features leading to the name lepre- morning due to late awakening and therefore a
chaunism. Most affected patients die in infancy, prolonged fast for the child). Alternatively, it
but if they survive, they may develop Leydig cell may present while the child has an intercurrent
hyperplasia, cystic ovaries, and hepatic fibrosis. illness that decreases dietary intake. The condi-
Pancreatic beta cell hyperplasia is characteristic. tion appears to result from a defect in mobiliza-
The Rabson–Mendenhall syndrome (#262190 tion of alanine for gluconeogenesis or from
ICD+ PINEAL HYPERPLASIA, INSULIN- decreased muscle mass, as muscles are the major
RESISTANT DIABETES MELLITUS, AND source of alanine. Affected patients with ketotic
SOMATIC ABNORMALITIES) combines fast- hypoglycemia not secondary to another diagno-
ing hypoglycemia with postprandial hyperglyce- sis tolerate an 18-h fast more poorly than do
mia and ultimately insulin-resistant diabetes typical children. This condition may be a more
mellitus as well as an enlarged pineal gland. exaggerated intolerance to fasting than normal,
Abnormal course facies are found, and enlarged as all of the features of ketotic hypoglycemia
clitoris or penis and pineal hypertrophy are will develop in a normal child who is fasted for
reported possibly with precocious puberty. Type an extended period. Normal children can tolerate
A insulin resistance (#610549 ICD+ DIABETES a fast lasting for 15 h by 1 year of age, 24 h by
MELLITUS, INSULIN-RESISTANT, WITH 1 year, and 36 h by 5 years without becoming
ACANTHOSIS NIGRICANS) leads to increased hypoglycemic.
growth and virilization as well as polycystic ova- The term ketotic hypoglycemia is quite gen-
ries. Reactive hypoglycemia has been described eral and includes many causes of hypoglycemia,
in this condition. including GH or GH resistance (see Chaps. 3 and
Munchausen by proxy or in the USA more 5), but does not include conditions involving
specifically, “factitious disorder imposed on increased insulin secretion. Adrenal failure,
another,” involves an adult caregiver who deliber- ACTH deficiency, and inappropriately rapid
ately makes a child sick. In one possible presenta- weaning after post-glucocorticoid therapy all
tion of this condition, a caregiver injects insulin into may lead to hypoglycemia (see Chap. 10). After
a child, which leads to unexplained hypoglycemia reviewing the history of the affected child, it
and/or seizure. Measurement of elevated serum may become clear that the child had neonatal
insulin during hypoglycemia with low C-peptide hypoglycemia, which apparently resolved after
indicates that the insulin is from exogenous regular feedings were started. However, the child
sources rather than the patient’s own pancreas may still maintain a tendency toward hypoglyce-
(C-peptide secretion only occurs with endoge- mia after prolonged fasting, which could be a
nous insulin secretion). Absence of hypoglyce- clue to a deficiency of GH or ACTH. Maple syrup
mia during scrupulous observation of the child, urine disease is a quite severe defect in the release
while eliminating the opportunity for anyone to of alanine from muscle, owing to a defect in
administer exogenous insulin, supports this diag- branched-chain amino acid catabolism (see ear-
nosis. Alternatively, a child being given sulfonyl- lier), which may be first diagnosed after infancy.
ureas will have elevated insulin secretion and Thus all ketotic hypoglycemia is not a benign
C-peptide secretion and will appear to have an variant, and some forms require medical rather
insulin-secreting condition. than only dietary management.
318 12 Hypoglycemia

Several types of GSD cause hypoglycemia, neogenesis is functional, the hypoglycemia is less
but hepatomegaly and poor growth might first severe, and many of the complications of GSD 1
bring affected infants to evaluation. GSD type 1 are absent. Hepatomegaly does, however, occur
(*232200 GLYCOGEN STORAGE DISEASE I because of increased glycogen stores. Frequent
at 17q21) is the result of a hepatic defect in feeding of glucose or cornstarch is the appropriate
glucose-6-phosphatase, the enzyme that converts therapy for most of these cases. However, some
glucose-6-phosphate to glucose in the last stage children have absence of amylo-1,6-glucosidase
of gluconeogenesis and glycogenolysis. The con- in the muscle as well and have weakness or even
dition is divided into types 1A and 1B (#232220 cardiac failure because of this defect; creatinine
GLYCOGEN STORAGE DISEASE IB at kinase is elevated in this condition.
11q23). Deficiency of this enzyme thus elimi- GSD type 6 (*232700 GLYCOGEN
nates both processes, which leads not only to STORAGE DISEASE VI at 14q21-q22) results
hypoglycemia but also to lactic acidosis and from a phosphorylase deficiency and GSD type 9
ketosis, hyperuricemia, hypophosphatemia, and (#306000 GLYCOGEN STORAGE DISEASE
abnormal platelet adhesiveness. The counterreg- IXa1; GSD9A1) from liver phosphorylase kinase
ulatory hormones rise during hypoglycemia, (PHK) deficiency; both conditions leading to
including glucagon, growth hormone, and corti- diminished glycogen breakdown to free glucose.
sol, produce hypertriglyceridemia and hypercho- Affected patients have hepatomegaly and possi-
lesterolemia, often severe. Presentation of bly hypoglycemia, but these are usually milder
hypoglycemia with lactic acidosis and ketosis conditions than GSD type 1.
may occur in the neonatal period or years later GSD 0 (#240600 GLYCOGEN STORAGE
after the development of characteristic growth DISEASE 0 at 12p12.2) is a rare condition caused
failure and hepatomegaly that is the result of gly- by hepatic glycogen synthetase deficiency, limit-
cogen and lipid deposition in the liver, although ing glycogen synthesis in the liver but not in the
liver function usually remains normal. The spleen muscle. Thus ketosis and hypoglycemia occur
is not enlarged, but the kidneys are enlarged with fasting, but elevated glucose is found after
because of glycogen deposition, and renal failure eating. The liver is of normal size. This condition
may develop, but this is not invariable. Type 1B resembles ketotic hypoglycemia in many aspects
GSD combines all the features characteristic of but has been reported in only a few individuals.
type 1A and, in addition, features neutropenia If a glycogen storage disease is strongly sus-
and infections that follow (oral and anal lesions pected, liver biopsy might be performed for
and chronic enteritis). Diagnosis is made by the definitive diagnosis.
administration of glucagon after meal (the fed Hereditary fructose intolerance is discussed
glucagon stimulation test) which in a normal above.
state raises glucose, but with these conditions, no Other causes of hypoglycemia include mal-
rise in glucose occurs but lactate rises. Genetic nutrition due to GI disease, diarrhea, or starva-
diagnoses are available. Treatment is frequent or tion, all of which lead to ketotic hypoglycemia.
constant administration of glucose, overnight by Alcohol ingestion or even significant skin expo-
nasogastric tube, as well as the ingestion of sure to alcohol can interfere with gluconeogen-
cornstarch to prolong the increase in blood esis since ethanol is metabolized by alcohol
sugar. The aim is to remedy the lack of available dehydrogenase in the liver to oxidize to acetal-
glucose and to suppress the counterregulatory dehyde and acetate which requires nicotinamide
hormone secretion that is responsible for many of adenine dinucleotide (NAD) as a cofactor limit-
the complications. ing gluconeogenesis; this limits the abundance
GSD type 3 (*232400 GLYCOGEN of NAD causing reduced gluconeogenesis and
STORAGE DISEASE III at 1p21) is the result of acidosis. Ackee fruit ingestion (Jamaican vom-
a deficiency in amylo-1,6-glucosidase, the deb- iting disease), Reye syndrome, or other causes
rancher enzyme for glycogenolysis; this is a of hepatic failure and the use of drugs that cause
milder condition than GSD type 1. Because gluco- hypoglycemia as a side effect, such as aspirin or
Hypoglycemia 319

oral hypoglycemic agents, also may precipitate Diagnosis of Hypoglycemia


hypoglycemia. Propranolol or other β-blocking
agents can cause hypoglycemia, especially after Medical history for a neonate must include the con-
a fast (e.g., when these agents are used to aug- dition of the mother and whether she had any type
ment the response to a GH-provocative test after of diabetes or intravenous glucose treatment. The
an overnight fast), and will also block adrener- onset of hypoglycemia in the fasted or fed state is
gic symptoms of hypoglycemia. Anorexia ner- of importance at any age: Infants will gently have
vosa and excessive exercise (not only in an hypoglycemia in a fasted state (although there are
HHF7) can cause hypoglycemia. As noted ear- exceptions as noted above), while older children
lier, many adults complain of what they call may have hypoglycemia in either situation.
hypoglycemia and often attribute symptoms in Questioning as to whether hypoglycemic agents
their children to the same condition. Symptoms are available in the house must be carried out.
generally occur a few hours after a meal, but A history of prior gastric surgery will also direct
when careful blood glucose measurements are the diagnosis.
obtained, low values are rarely found and even Physical examination will aim to discover the
more rarely match the onset of symptoms. physical findings of an infant of a diabetic mother,
Although a 5-h glucose tolerance test with mea- small-for-gestational-age infant, and findings of
surement of glucose and insulin might be con- Beckwith–Wiedemann syndrome. Midline
sidered, in 25 % of individuals, the blood sugar defects and microphallus in boys (penis less than
decreases during an oral glucose tolerance test 2 cm in length) are indicative of hypopituitarism.
(OGTT) as 10 % of normal adults experience a A large liver is compatible with GSD.
blood sugar below 50 mg/dL on the third to fifth Diagnosis of hypoglycemia must involve
hours of a glucose tolerance test, more if CH2O measuring blood sugar rather than just assuming
loading is not provided for 3 days beforehand, that it is low or relying on a finger-stick glucom-
thus falsely suggesting the diagnosis of hypo- eter measurement (Fig. 12.3). The method of
glycemia. Most experts do not support the exis- measurement of glucose deserves attention due
tence of the condition known as reactive to several practical issues. As stated above,
hypoglycemia. Of course, it is harmless to mod- ,plasma glucose is 10–15 % lower than blood
ify the diet of most children to eat breakfast, glucose. Blood samples for a metabolic panel (in
avoid concentrated sugar, and consider a midaft- any red top tube) that sit around at room tem-
ernoon or a midmorning snack (again not exces- perature for hours without fluoride in the tube
sively sweetened or excessively caloric in this will have a decrease in blood sugar of 5–7 mg/dL
epidemic of obesity), a measure often suggested per hour, and the resulting difference over 2 h
by those who support the diagnosis of reactive can blur the distinction between normal blood
hypoglycemia. If frequent feedings are advised sugar and hypoglycemia. Tubes containing fluo-
to avoid factitious hypoglycemia, excessive ride (grey top), which stops the metabolism of
caloric intake and obesity might be the result. If glucose by blood cells and allows the storage of
in doubt, rather than supporting an inappropriate blood for hours without reducing the glucose
diagnosis, one might discuss with the family that concentration, are the best method of collection.
since simple measures of dietary manipulation Finger-stick methods of assessing blood glucose
relieve the symptoms, just follow them without are available by glucometer, but these must be
labeling the child. Further, it is important to real- considered as a guide and not definitive glucose
ize that autonomic dysfunction leading to pos- results. Inadequate sampling or measurements or
tural hypotension and other symptoms may be out-of-date strips will affect the results.
the true cause of the symptoms labeled as reac- Glucometers are designed for the management
tive hypoglycemia. Adequate hydration is usu- of diabetes, in whom elevated values are fre-
ally suggested as the best treatment for those quent, so they do not reliably measure lower val-
conditions. ues. A blood or plasma glucose reading at a
320 12 Hypoglycemia

Diagnostic Schema From Results of Critical Sample


Hypoglycemia

reduced blood sugar (generally less than 50 mg/dl)

historical features such as history of diabetes mellitus under treatment?


No Yes

obtain critical blood sample with glucose, insulin, GH, cortisol, ketone, lactate, pyruvate and other tests consider excess insulin or oral hypoglycemic agents
listed on Table 11.2 IF THIS SAMPLING IS POSSIBLE WITHOUT ENDANGERING THE PATIENT

high for glucose value (FFA will be low suppressed as expected in hypoglycemia (FFA will be high
and elevation of glucose occurs after insulin and no elevation of glucose occurs after glucagon)
glucagon)
ketones
positive negative
c peptide normal
high low low GH elevated deficient FFA transport
ACYL-CoA DEHYDROGENASE,
neonatal period after
GH deficiency cortisol MEDIUM CHAIN AD DEFICIENCY
neonatal
period low normally elevated
infant of ACTH or cortisol deficiency lactate and alanine
diabetic mother exogenous insulin administration low high
Munchausen's by proxy
liver size defect in transient hypoglycemia of infancy
enlarged normal gluconeogenesis
persistent hyperinsulinemic hypoglycemia
islet cell adenoma abnormal glycogenolysis "ketotic early onset after
Beckwith Weideman syndrome phosphorylase deficiency hypoglycemia" onset neonatal/infant period
MEN type I debrancher enzyme deficency
sulfonurea exposure Glycogen storage disease types 1,3,0
fructose 1-6 diphosphatase deficiency alcohol or salicylate intake
hereditary fructose intolerance or Reye syndrome
PEPCK
PC
galactosemia
Glycogen storage disease types 1,3,9,0 eg., glucose 6 phosphase deficiency

Fig. 12.3 Flow chart of the diagnosis of hypoglycemia

licensed laboratory must confirm low blood glu- defect in hepatic gluconeogenesis such as glucose
cose values below 55–60 mg/dL. Nonetheless, if 6-phosphatase or fructose 1,6-diphosphatase defi-
used appropriately, with reagents or test strips ciency if ketones are absent or in older children
that are not outdated, glucometers can be a use- suggests ethanol ingestion), pyruvate (low pyru-
ful adjunct to diagnosis and follow-up for ade- vate suggests difficulty in mobilizing substrate for
quacy of treatment, as long as true laboratory gluconeogenesis) determinations, C peptide (to
values are used as well. Methods measuring separate endogenous versus exogenous insulin
reducing substances, such as Clinitest, will mea- sources), ammonia (elevated in certain hyperinsu-
sure other substances, such as galactose, whereas linemic conditions and along with elevated trans-
those using a glucose oxidase technique will be aminases in fatty acid oxidation defects), amino
specific for glucose. acids, free and bound carnitine (to determine if
there is a defect in mitochondrial beta oxidation),
The Critical Sample and, if blood volume allows, free fatty acid (which
When a child has hypoglycemia, a blood sample are elevated in fatty acid oxidation defects such as
should be obtained immediately so that determi- medium-chain AD deficiency but decreased with
nations crucial to the diagnosis can be ordered; increased insulin secretion) determinations, and
this is the “critical” blood sample (Table 12.2) that extra serum should be frozen for future determi-
is the mainstay of diagnosis. Thus, if BS is nations in case another area of diagnostic inquiry
<55 mg/dL, obtain blood for glucose (a true labo- seems more likely after the initial results are back
ratory blood glucose rather than a glucometer (such as IGF-1, IGF-2 which may be produced by
value), insulin (a value of insulin/glucose greater tumors, IGF BP1 which is regulated by insulin
than 0.3 suggests hyperinsulinism if serum glu- rather than growth hormone and is decreased with
cose is less than 55 mg/dL, but small increases in excessive insulin secretion.) Urine should be
insulin secretion below the detection limit of the obtained for determination of ketonuria at the
laboratory are still compatible with hyperinsulin- same time. The presence of ketone bodies in the
ism), serum ketones (see below about ketosis), urine or blood at the time of hypoglycemia will
growth hormone and cortisol (both should rise reflect the insulin secretory state; ketotic hypogly-
with hypoglycemia), lactate (elevation suggests a cemia is virtually incompatible with hyperinsulin-
Hypoglycemia 321

Table 12.2 The critical blood sample (or other tube with fluoride) tube initially to stop
To be obtained if BS is <50 mg/dL metabolism of glucose or if the blood cells are
Glucose separated from plasma or serum right away, the
Insulin glucose value is more accurate.
Ketones After the critical blood sample is drawn, glu-
Growth hormone cose is given to increase the blood sugar; the hypo-
Cortisol glycemic episode should not be dangerously
Lactate prolonged just to obtain blood for the diagnostic
Alanine sample, of course. Anticipating the possibility that
Pyruvate hypoglycemia may occur in a predictable situation
Also urine organic acids will lessen the effort later required for the diagno-
If possible: sis and treatment of the condition. Thus patients
Free fatty acids with stressful deliveries, with prematurity, or with
C peptide IUGR or, conversely, infants with large body size
IGF-1 suspected to be the offspring of a mother with dia-
IGF-11 betes mellitus should have blood sugar monitoring
IGF BP1 at frequent intervals during the first 24 h after birth.
Ammonia
In neonates, fasting hypoglycemia most often
Amino acids
occurs, but rarely postprandial hypoglycemia
Free and bound carnitine
presents. In older patients, the pattern of hypo-
Save as much serum/plasma as possible for future
determinations
glycemia is of extreme importance in the differ-
ential diagnosis. Fasting hypoglycemia more
IGF insulin-like growth factor
likely will indicate a defect in glucose production
or release, such as a defect in gluconeogenesis or
ism but occurs in GH or cortisol deficiency, but a GSD, whereas postprandial (or true reactive)
nonketotic hypoglycemia increases the likelihood hypoglycemia suggests a hyperinsulinemic state;
that insulin may be the etiologic agent in the con- overlap occurs, however, as hyperinsulinism will
dition. Ketosis-accompanying hypoglycemia will cause fasting hypoglycemia as well (Table 12.1).
also occur in ketotic hypoglycemia or GSD type If the critical initial sample is not collected, sev-
III. Hypoglycemia is diagnosed only if a true eral steps should follow. If the hypoglycemia is not
serum/plasma sugar is low or if a glucometer indi- so severe and urgent, frequent measurement of
cates a low blood sugar that is later confirmed blood glucose is made during a 24-h period, before
when the laboratory determination returns; if only each meal, and at 1, 2, and 3 h after one or more
symptoms that suggest hypoglycemia are meals and also measured in the middle of the
reported, a diagnosis of true hypoglycemia is not night; this might be called a “nothing tolerance
appropriate until confirmed. test.” If the blood sugar is decreased below 55 mg/
The aim of management of a hypoglycemic dL in any of these samples, as reflected by a finger-
child is to prevent the recurrence of hypoglyce- stick glucose method by glucometer, the full set of
mia; if the critical blood sample is missed on pre- critical value tests noted earlier is performed along
sentation, it may require long periods of with a true laboratory glucose determination, if
observation before a recurrence allows the possible, before glucose is given to increase the
diagnostician to have another chance at diagnosis blood sugar concentration. The critical sample
with a critical blood sample during another hypo- including an insulin determination should be mea-
glycemic event. Remember that the critical sam- sured contemporaneously with a low glucose con-
ple must be processed quickly so that glucose is centration, not sequentially.
not decreased by the metabolic activity of blood If no episode of spontaneous hypoglycemia
cells in a collection tube in case the blood was occurs, an observed fast should be performed
collected without fluoride additives; if the glu- after a high-carbohydrate diet is given for 3 days
cose determination is separated into a grey top before the beginning of the fast. If the child’s
322 12 Hypoglycemia

carbohydrate stores are not repleted by the high- no increase in glucose will be seen in GSD type 1
carbohydrate diet, specious hypoglycemia may (glucose-6-phosphatase deficiency), whereas in
occur. The fast should start after a usual over- ketotic hypoglycemia or GSD type 3, a normal
night period of sleep (if the child usually goes response will occur in the fed state and a poor
without food at night), so that 10–12 h of the rou- response after a fast. The fructose tolerance test is
tine fast starts at 7:00–8:00 a.m., a time when the no longer recommended to evaluate hereditary
full hospital staff should be present to handle fructose intolerance or 1,6-diphosphatase defi-
potential complications. Finger-stick glucose ciency as genetic tests are now available.
determinations by glucometer are made every 2 h Some tests cannot be recommended without
until a tendency is noted for a decrease in glu- prior experience. They are the leucine (orally, dis-
cose. All urine samples are analyzed for ketone solved in a slurry of CO2-free water administered
bodies as in ketotic hypoglycemia the appearance through a stomach tube, at 150 mg/kg, or intrave-
of ketonuria will precede hypoglycemia. At the nously, 75 mg/kg, and blood obtained for glucose
time that glucose decreases on the glucometer, a and insulin at 5, 10, 20, 30, 45, 60, 90, and 120 min
true laboratory glucose measurement is made at or every 10 min for an hour, respectively) or the
the same time as the finger-stick sample and the tolbutamide tolerance test (20–30 mg/kg up to 1 g
critical sample obtained. If the fast precipitates intravenously and blood sampled at 5, 10, 20, 30,
no decrease in glucose by 5:00 p.m. (after 45, 60, 90, and 120 min) to determine if insulin
20–22 h), the test can be continued for a full 24 h secretion is inappropriately high. The tests may be
in an infant or even 36 h in the older child if ade- dangerous, as severe hypoglycemia may be pre-
quate staff is available to monitor the child safely. cipitated by these insulin secretagogues.
If the glucose concentration decreases to less The glucose tolerance test (GTT) is rarely
than 55 mg/dL, glucagon is given at a dose of indicated, but if performed to rule out reactive
0.02–0.03 mg/kg to a maximum of 1 mg, and hypoglycemia, it is important to carry out several
after 10 min, blood glucose, insulin, lactate, and important procedures to ensure that false results
pyruvate are measured to see if the child has ade- do not confuse the issue: 3 days of high-
quate glycogen stores or if there is an abnormal- carbohydrate diet must be ingested or the response
ity in mobilization of glucose. Glucagon should to the oral glucose load will be falsely abnormal,
not, however, be given if hepatomegaly is pres- even if the patient is normal (this type of prepara-
ent, as it might worsen metabolic acidosis caused tion also is important for the other tolerance tests
by a possible gluconeogenic defect. The fast is listed earlier); an insulin determination should
usually stopped by 24 (or 36) as hours, and if no accompany each glucose measurement; the test
hypoglycemia is demonstrated, it is at least should extend for 5 h to look for late hypoglyce-
apparent that the child can tolerate a 24-h fast at mia that would be missed on a shorter test; and
home; such a long fast is not, of course, recom- the dose of glucose is 1.75 g/kg, as a 20 % solu-
mended, but a good margin of error exists if the tion. The symptoms manifested by the patient
child is fed on a reasonable schedule. It should be should be recorded during the test so that any low
clear that if the initial critical sample is obtained blood sugar determinations can be matched with
appropriately, this complex diagnostic plan might symptoms of hypoglycemia, making an organic
often be eliminated altogether. abnormality more likely. Just as important as
Various tolerance tests are used in the differ- determining that a low blood sugar is associated
ential diagnosis of hypoglycemia. The glucagon with the symptoms is the elimination of the
tolerance test (0.3 mg/kg body weight (maximum symptoms with an increase in blood sugar.
dose 500 μg) administration of glucagon intrave-
nously or intramuscularly, with blood glucose,
FFAs, ketones, insulin, and GH determinations Treatment of Hypoglycemia
obtained at 0, 5, 15, and 15 min thereafter for 2 h)
determines whether glycogen can be broken Treatment of hypoglycemia may be a short-term
down to supply glucose; in the fed or fasting state, initial treatment to correct the specific event or
Hypoglycemia 323

long-term treatment to ensure continuation of a Table 12.3 Treatment of hypoglycemia


normal state: prolonged treatment is best aimed Acute
to achieve a blood sugar value greater than If conscious Oral sugar, cake
60 mg/dL at all times and normal values of frosting or glucose gel,
70–120 mg/dL at least most of the time (Table 12.3; 4 oz juice
see Chap. 15 for discussion of emergency treat- If unconscious i.v. Dextrose bolus,
25 % at 1 mL/kg
ment of hypoglycemia). The initial treatment of
If diabetic or if no i.m. Glucagon, 30 μg/kg
hypoglycemia must be the administration of glu- evidence of GSD and to a maximum of 1 mg
cose sufficient to bring the blood glucose to ade- hepatomegaly
quate levels and to return neurologic function to Chronic
normal but not excessive glucose to cause severely Ketotic hypoglycemia Frequent feedings
increased osmolality. In a child who is only GSD 1 Frequent feedings and
moderately symptomatic, oral glucose is adequate; cornstarch
table sugar, honey (only if the child is older than Hereditary fructose Avoidance of fructose
intolerance
1 year or better yet 2 years and not at risk for infant
Galactosemia Avoidance of galactose
botulism), or jelly can be administered orally or to
Hyperinsulinism Diazoxide, octreotide,
the child’s buccal mucosa if the child is too disori- or pancreatectomy
ented to swallow. For more severe hypoglycemia, Carnitine deficiency Carnitine
intravenous glucose is necessary, but if dextrose MCAD Avoid fasting
in higher concentration (dextrose vials are sup- Long-chain fatty acid Medium-chain fatty
plied as 50 %) is administered, a risk of hyperos- abnormalities acids
molality exists, especially in infants and neonates. GSD glycogen storage disease, MCAD medium-chain
Intravenous D10W is preferable to more concen- acyl CoA dehydrogenase
trated preparation in a dose of 2 mL/kg which is
0.2 g/kg of dextrose over 1 min. Alternatively must be present in the infusion to avoid hypona-
glucose given in 25 % dextrose as a bolus of tremia developing as the hypoglycemia is
1 mL/kg is reasonable. Once the blood sugar repaired. It is useful to calculate the amount of
increases, the child should be observed longer; if glucose in terms of milligrams per kilogram per
the sugar again decreases, a bolus of 5 mL per minute to determine the severity of the hypogly-
kilogram of 10 % dextrose is administered, and if cemia. If 6–8 mg per kilogram per minute of dex-
necessary, an infusion of dextrose, 5–10 %, trose will support the child’s glucose,
should be started; appropriate sodium chloride hyperinsulinism is virtually eliminated.

( Dextrose Concentration in g/dL expressed as an integer) ´ ( mL/hour)


Glucose Infusion Rate (or GIR) in mg/kg/minute =
( Weight in kg ) ´ (6))

If more than 10–12 mg/kg/min of glucose or more The treatment of several specific diagnoses was
is required repeatedly, the child has severe hypo- presented earlier. Dietary management is possi-
glycemia, most likely due to hyperinsulinism. ble in some conditions.
Abrupt discontinuation of dextrose support may Frequent carbohydrate feeding is the manage-
lead to rebound hypoglycemia after the normal ment for uncomplicated ketotic hypoglycemia.
increase in insulin secretion caused by dextrose GSD type 1, a gluconeogenic defect, is treated
administration. This rebound condition should not with frequent carbohydrate feedings, but overnight
be confused with bona fide hyperinsulinism. As blood glucose is supported by uncooked cornstarch
transient hypoglycemia resolves, weaning off i.v. preparation or continuous nasogastric feedings.
dextrose is followed by frequent oral administra- Hereditary fructose intolerance or fructose-
tion of glucose-containing substances, as the child 1,6-bisphosphatase deficiency mandates avoid-
can tolerate under careful observation. ance of fructose and sorbitol.
324 12 Hypoglycemia

Galactosemia mandates avoidance of galac- Suggested Reading


tose ingestion.
Fatty oxidation defects, including MCAD, 1. Langdon DR, Stanley CA, Sperling MA. Chapter 21 -
Hypoglycemia in the toddler and child. In: Sperling
may respond to a low-fat diet and avoidance of MA, editor. Pediatric endocrinology. 4th ed.
fasting that might cause lipid breakdown. Philadelphia, PA: Elsevier; 2014. p. 920–955.e921.
In defects of long-chain fatty acid metabolism, 2. De León DD, Thornton PS, Stanley CA, Sperling
medium-chain triglycerides are administered, and MA. Chapter 6 - Hypoglycemia in the newborn and
infant. In: Sperling MA, editor. Pediatric endocrinol-
carbohydrates or glucose given during fasting or ogy. 4th ed. Philadelphia, PA: Elsevier; 2014. p. 157–
stress that might lead to lipid breakdown. 185.e152.
Carnitine is administered for primary or sec- 3. Ly TT, Maahs DM, Rewers A, Dunger D, Oduwole A,
ondary carnitine deficiency. Jones TW. Assessment and management of
hypoglycemia in children and adolescents with diabe-
Other measures are used for the management of tes. Pediatr Diabetes. 2014;15 Suppl 20:180–92.
severe hypoglycemia. Diazoxide, a benzothiadia- 4. Park E, Pearson NM, Pillow MT, Toledo A. Neonatal
zine hypotensive agent, in doses of 5–20 mg/kg/ endocrine emergencies: a primer for the emergency
day divided into a dose every 6 h, will suppress physician. Emerg Med Clin North Am. 2014;32(2):
421–35.
insulin secretion in some cases of hyperinsulinism, 5. Tin W. Defining neonatal hypoglycaemia: a continuing
as noted earlier, but will not usually be effective in debate. Semin Fetal Neonatal Med. 2014;19(1):27–32.
β-cell adenomas. A lack of response to diazoxide is 6. Snider KE, Becker S, Boyajian L, Shyng SL,
not diagnostic but does suggest the presence of a MacMullen C, Hughes N, Ganapathy K, Bhatti T,
Stanley CA, Ganguly A. Genotype and phenotype
β-cell adenoma or one of the hyperinsulinemic correlations in 417 children with congenital hyperin-
states discussed earlier that will require surgical sulinism. J Clin Endocrinol Metab. 2013;98(2):
removal. Diazoxide has several possible side E355–63.
effects including hirsutism, water and sodium 7. Lang TF, Hussain K. Pediatric Hypoglycemia. Adv
Clin Chem. 2014;63:211–45.
retention (may be opposed by the use of hydrochlo- 8. UpToDate Neonatal hypoglycemia Author See Wai
rothiazide which does not itself cause hypoglyce- Chan, MD, MPH
mia mild hyperuricemia, advanced bone age, and 9. Sunehag A and Haymond MW. Approach to hypogly-
decreased immunoglobulin G and neutrophil num- cemia in infants and children. http://www.uptodate.
com/contents/approach-to-hypoglycemia-in-infantsand-
bers. Octreotide is a synthetic long-acting soma- children.
tostatin analogue that will suppress insulin 10. Sunehag A and Haymond MW. Causes of hypoglyce-
secretion in some cases. Dosing starts at 5 μg/kg mia in infants and children. http://www.uptodate.
subcutaneously, titrating the dose to 40 μg/kg/24 h, com/contents/causes-of-hypoglycemia-in-infantsand-
children.
divided into six doses per day. Tachyphylaxis is a 11. Rozance PJ. Pathogenesis, screening, and diagnosis
possible side effect, as is steatorrhea. of neonatal hypoglycemia. http://www.uptodate.com/
Pancreatectomy is recommended in cases of contents/pathogenesis-screening-and-diagnosisof-
hyperinsulinism resistant to diet or medications neonatal-hypoglycemia.
12. Rozance PJ. Management and outcome of neonatal
and should not be delayed if no other treatment hypoglycemia. http://www.uptodate.com/contents/
successfully supports the blood sugar. management-and-outcome-of-neonatal-hypoglycemia.
Glucocorticoids are also used to increase blood 13. Sunehag A and Haymond MW. Pathogenesis, clinical
sugar but should not be used for long-term replace- features, and diagnosis of persistent hyperinsulinemic
hypoglycemia of infancy. http://www.uptodate. com/
ment, except in cases of cortisol or ACTH defi- contents/pathogenesis-clinical-features-anddiagnosis-
ciency, and then only in replacement doses. GH of-persistent-hyperinsulinemic-hypoglycemia-of-
therapy is offered in cases of GH deficiency. The infancy.
most important part of therapy is the rapid institu- 14. Sunehag A and Haymond MW. Treatment and com-
plications of persistent hyperinsulinemic hypoglyce-
tion of a treatment designed to increase blood mia of infancy. http://www.uptodate.com/contents/
sugar and guard against severe and repetitive hypo- treatment-and-complications-of-persistent-hyperin-
glycemia and thereby to salvage mental function. sulinemic-hypoglycemia-of-infancy.
Obesity
13

young years, American Indians and Hispanics in


Definition the first years have prevalence of obesity close to
18 %. In 2011–2012, 17.7 % of 6- to 11-year-olds
Obesity is the most prevalent nutritional disorder of and 20.5 % of 12- to 19-year-olds were obese.
children in the USA, a remarkable change from the Obesity is higher if the parents have not com-
prominent place of malnutrition due to deficient pleted college in most groups. Waist circumfer-
caloric intake in childhood in the past. Surgeons ence serves as a reflection of visceral adiposity,
General have designated childhood obesity as an and waist circumference has increased faster than
epidemic in the USA. A threefold increase in the BMI over the last decade in children but has also
prevalence of a body mass index (BMI) at or plateaued since 2004.
greater than the 95th percentile for age and gender Overweight is defined as a BMI ≥85th for
in children aged 6–11 years occurred between the ­percentile age and gender and less than 95th per-
National Health and Nutrition Evaluation II (1976– centile. A child less than 2 years of age is obese
1980) and 2004. This trend in most groups has lev- if the weight for recumbent length is greater than
eled off since 2004. At ages 2–19, the prevalence is or equal to the 97.7th percentile of WHO growth
about 17 % (or 12.7 million). standards (charts on CDC website).
Obesity is a striking example of health dispari- A new classification of severe extreme obesity
ties. The prevalence is higher among Hispanic applies to children whose BMI is at or above the
(22.4 %) and non-Hispanic black youth (20.2 %) 99th percentile due to the more severe morbidity
than non-Hispanic white youth (14.1 %) and non-­ and mortality found with this classification. More
Hispanic Asian youth (8.6 %). Obesity in the recently, guidelines recommended the diagnosis of
Asian population in the ages between 5 and 19 severe obesity if a BMI is above 120 % of the 95th
years has increased to 127 % between 1992 and percentile, a BMI is +2.33 standard deviations
2001 which is surprising in a population notable above the mean, or a BMI is greater than 35 kg/m2
for its previous leanness. Obesity increases with (Fig. 13.1). Unfortunately, the classification of
age, but young children are already demonstrating severe obesity is the most rapidly growing classifi-
obesity with a high prevalence: in 2003–2004, the cation in childhood and adolescence and presently
prevalence of obesity in 2–5-year-olds was includes 4–6 % of the population at these ages. We
13.9 % (the prevalence dropped to 8.4 % in 2011– do not use the term “morbid obesity.” We speak of
2012, but there is controversy if this unexpected healthy habits in diet and activity in communica-
drop in the face of no change in other age groups tion with the family and patient rather than the term
across the same time is correct and long lasting or obesity, although “obesity” and “overweight” are
whether it is a short-term variation). Already at used in this chapter as medical terms.

© Springer International Publishing Switzerland 2016 325


D.M. Styne, Pediatric Endocrinology, DOI 10.1007/978-3-319-18371-8_13
326 13 Obesity

2 to 20 years: Boys NAME


Body mass index-for-age percentiles RECORD #

Date Age Weight Stature BMI* Comments


BMI

35

34

33

32

31

30
95
29

BMI 28
90
27 27
26 85 26
25 25
75
24 24

23 23
50
22 22

21 21
25
20 20
10
19 19
5
18 18

17 17

16 16

15 15

14 14

13 13

12 12

kg/m
2
AGE (YEARS) kg/m
2

2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20

Published May 30, 2000 (modified 10/16/00).


SOURCE: Developed by the National Center for Health Statistics in collaboration with
the National Center for Chronic Disease Prevention and Health Promotion (2000).
http://www.cdc.gov/growthcharts

Fig. 13.1 (a) Body mass index-for-age percentiles for girls aged 2–20 years (from http://www.cdc.gov/growthcharts/).
(b) Body mass index-for-­age percentiles for boys aged 2–20 years (from http://www.cdc.gov/growthcharts/)
Definition 327

2 to 20 years: Girls NAME


Body mass index-for-age percentiles RECORD #

Date Age Weight Stature BMI* Comments


BMI

35

97
34

33

32

95
31

30

29

BMI 28
90

27 27
26 85 26
25 25

24 75 24

23 23

22 22
50
21 21

20 20
25
19 19
10
18 18
3
17 17

16 16

15 15

14 14

13 13

12 12

kg/m
2
AGE (YEARS) kg/m
2

2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20

Published May 30, 2000 (modified 10/16/00).


SOURCE: Developed by the National Center for Health Statistics in collaboration with
the National Center for Chronic Disease Prevention and Health Promotion (2000).
http://www.cdc.gov/growthcharts

Fig. 13.1 (continued)
328 13 Obesity

The disparities in obesity will translate into the (highly accurate but expensive and requires
disparities in morbidity and mortality in adult life. exposure to radiation). To simplify the issue,
The economic cost of the epidemic is the sum of anthropologic methods are used in the office set-
medical care costs, loss of productivity, and of ting to express percentage of body fat. In the past,
course the tragedy of chronic disease in an indi- percentile of weight for age may have been used
vidual; the Brookings Institute estimates that if all to reflect the percentage of body fat, but this is
the 12.7 million obese US youth became obese not helpful in childhood, as it does not take into
adults the lifetime cost will exceed $1.1 trillion. consideration the height of the child. Percentile
Obesity is even affecting national security since weight for height is a better indication, but does
anywhere between 25 and 75 % of youth applying not adequately express the relative amount of fat
to the various uniformed services are refused in the individual, because increased muscle tis-
because they are not in optimal health. sue will incorrectly increase the weight for
It is of course a mathematical impossibility height. BMI was adopted as the international
for 17 % of a population to be above the 95th standard of adiposity as it is considered to reflect
percentile as only 5 % should be above that level. the percentage of body weight that is fat gener-
Presently the CDC is setting normal values for ally reliably, and BMI correlates with morbidity
weight in children by the use of statistics from and mortality in adults. BMI is known to be more
decades predating the obesity epidemic. The accurate in adults than in children, but it is now
CDC weight charts are based upon more than the standard of clinical practice in children as
three-decade-old data although the CDC height well. BMI should be calculated and interpreted
charts derive from more recent data. for every child at every office visit. Electronic
Areas of the world previously suffering from medical record systems often calculate the BMI
malnutrition are now also manifesting an increas- automatically, eliminating the need for manual
ing prevalence of obesity, with the introduction calculation. If EMR does not have this capability,
of a Western (mainly US style) diet and lifestyle. there are numerous websites to easily input
Type 2 diabetes mellitus is likewise widespread height and weight and find the BMI percentage
in these same regions of the world and children after input of age and gender as well. Below is
figure prominently in these trends. The preva- the equation for the calculation of BMI:
lence of diabetes is predicted to increase by 51 %
worldwide by 2030, with the majority of that BMI in kg / m 2 = weight in kg / height in meters 2
increase occurring in the developing world.
The definition of obesity is an excess of adi- or
pose tissue, not just an increase in weight. The
703 ´ Weight in pounds ¸ ( Height in inches )
2
adverse metabolic and psychological effects of
this adipose tissue are of paramount importance,
but the psychological effects of obesity are sub- The percentile of BMI for age changes by
stantial as well. Obesity can be expressed as the age in children, so no simple number defines
percentage of the body consisting of fat. Many obesity at all ages, as in adults where a BMI >25
techniques of measurement of body composition is overweight, >30 is obese, etc. A chart of BMI
are available: measurement of subscapular skin versus age from the Centers for Disease Control
thickness (requires special training and is diffi- (CDC) should be used to evaluate all children
cult to reproduce in a clinical office), underwater (Fig. 13.1).
weighing (requires a tank of water and other Absolute fat mass is relatively constant in
rarely available materials), bioelectric imped- both males and females until just prior to
ance (easy to perform but sometimes difficult to puberty. During puberty, girls begin gaining sig-
standardize at a child’s age and not accurate in nificantly more fat mass than males and by the
the extreme cases of obesity), dual X-ray absorp- end of puberty have about 5–6 kg more absolute
tiometry (DXA, which is accurate but expen- fat mass, representing an average gain of 1.14 kg
sive), or computed tomography (CT) scans per year.
Etiology of Obesity 329

The Basic Approach to Childhood Obesity adiposity rebound. The adiposity rebound is
noted on the BMI chart as an upward curve fol-
Prevention
Always Primary lowing the early childhood nadir at about 5–6
years, earlier in girls. An early adiposity rebound
before 4 years or younger increases the likeli-
hood of the development of obesity and meta-
BMI > 85th percentile?
bolic syndrome.
History
Clinically, a child who is just above the 85th
Risk factors? percentile may not appear to have excessive adi-
Symptoms of pose tissue at casual glance, while in those above
comorbidities?
the 95th percentile, excessive adipose tissue
should be more obvious. It is of interest to check
Physical
Comorbidities?
the BMI of the child who appears to be “thin” in
the office setting. In the absence of disease, you
Laboratory studies based upon
might find that the BMI is actually at the 50th
findings and BMI % percentile; we are so used to seeing overweight
and children with obesity in America that an
Determine need for weight maintenance or weight loss average child may seem abnormally thin.

Evaluate stage of readiness to change


Etiology of Obesity
Use motivational interviewing to
determine appropriate targets for
change behavior
 enetic Factors and the CNS Control
G
of Appetite
Maintenance!
Strong genetic and environmental factors affect
Fig. 13.2  The Basic Approach to Childhood Obesity obesity, and both factors play a role in the obesity
epidemic. Thus obesity should not simply be
considered a condition of overeating and
­underexercising, although both activities play a
Males do not significantly change their abso- role in the etiology, and both activities must be
lute fat mass during their pubertal years. In males, changed to control obesity; it is inappropriate and
the net effect of these changes is a decreasing certainly not clinically helpful to consider obesity
percent body fat throughout puberty to an adult as a simple manner of lack of an individual’s self-­
value of approximately 13 %. In females, the net control. Between 40 and 80 % of the variance in
effect of a plateau in fat-free mass and increased BMI is determined by genetic factors; greater sim-
fat mass results in an increasing percent body fat ilarity is found in the BMIs of monozygotic twins
which averages 25 % in adulthood. than in those of dizygotic twins, and the BMI of an
Adults have increased morbidity and mortality adopted child relates far better to the BMI of the
when their BMI increases to more than 25 (clas- biologic parents than to those of the adoptive par-
sified as overweight), which is the 85th percentile ents with whom the child was raised.
of BMI for adults. By extension, a BMI for chil- In spite of this, the loci that are identified to
dren that is at or above the 85th percentile for age affect BMI to date have not shown major effects.
and gender is considered overweight, whereas at Epigenetics refers to the modification of gene
or above the 95th percentile BMI for age, a child expression by environmental influences, and this
is considered to be obese (Figs. 13.1 and 13.2). process is certainly demonstrated in the origins
Normally, an increase in the percentage of of obesity; when pregnant women have experi-
body fat is noted at mid-childhood, called the enced starvation, for example, during famine in
330 13 Obesity

World War II, their offspring often have an Parenthetically, the anti-inflammatory cyto-
increased tendency toward obesity later in life. kine adiponectin decreases in obesity with insu-
Gestational effects are important as a macroso- lin resistance and rises with a healthy weight.
mic infant has a higher likelihood of becoming Adiponectin can be seen as an inverse barometer
an obese 10-year-old with insulin resistance. of the metabolic consequences of obesity.
Further, smaller-birth-­weight infants owing to a Ghrelin is a peptide that is released from the
variety of conditions have a tendency to develop stomach, colon, and small intestine to act as a
insulin resistance later in life and obesity. Parental signal to the hypothalamus to increase feeding
effects may be seen in the case of a child with one (orexigenic). Ghrelin acts as a growth hormone
obese parent who has a fivefold increase in the secretagogue through the pituitary gland growth
likelihood of becoming an obese adult, and a hormone secretagogue receptor (GHS-R) in the
child with two obese parents who has a 12-fold arcuate nucleus.
increased risk. Peptide YY (PYY) and glucagon-like peptide
Single-gene defects account for less than 7 % 1 (GLP-1) are produced from the intestinal L
of severe obesity but can exert remarkable effects cells (or enteroendocrine cell). The PYY crosses
(see below). The study of individuals with single-­ the blood–brain barrier to promote satiety. GLP-1
gene mutations and animal models of obesity has has numerous effects including inhibition of gas-
led to increasing knowledge about the regulation tric emptying, promotion of insulin secretion
of appetite. after eating, and promotion of satiety. The effect
Several hormones from distal sites act on the of GLP-1 is utilized in medications known as
CNS to regulate appetite. Leptin is a peptide hor- incretins in the treatment of type 2 DM, while
mone produced by adipocytes, and serum levels inhibition of the enzymes that metabolize GLP-1,
of leptin directly correlate with the amount of dipeptidyl peptidase-4, or DPP-4 enhances
adipocytes. Leptin crosses the blood–brain bar- endogenous GLP-1.
rier and attaches to a receptor on the melanocor- Insulin plays an important role in the metabo-
tin cells in the arcuate nucleus of the ventromedial lism of carbohydrate protein and fat as described
hypothalamus to decrease food intake in an in Chaps. 11 and 12.
anorexigenic effect. Other hormones are produced in the CNS to
Loss-of-function mutations in the leptin gene regulate eating behavior. Melanocortin cells pro-
or the leptin receptor gene lead to early-onset duce proopiomelanocortin (POMC) which in
progressive obesity and extreme hyperphagia. turn is cleaved into various peptides depending
Leptin deficiency also eliminates the onset and upon the tissue where processing occurs. One
progression of puberty. However, when leptin is product of POMC is the melanocyte-stimulating
administered to leptin-deficient subjects, appetite hormone (MSH) which is an anorexigenic hor-
decreases to normal levels, weight loss proceeds mone (POMC is further described in Chap. 10).
while activity levels increase, and if they are of MSH interacts with the MC4 receptor (MC4R)
appropriate age, puberty progresses. to decrease appetite and the MC3 receptors
Although the rare mutation in genes for leptin (MC3R) to increase feeding efficiency. Leptin
or for leptin receptors in human beings (only a decreases the production of two factors which
few cases of loss-of-function mutations are increase caloric intake: (1) agouti-related protein
reported) leads to early-onset, severe obesity. (AGRP) which blocks the MSH stimulation of
Leptin levels are high in most obese human the MC4R and MC3R receptors leading to
beings. Because appetite in obese individuals is decreased satiety and (2) neuropeptide Y which
not suppressed by these values of leptin, a rela- strongly stimulates appetite. The melanin-con-
tive leptin resistance occurs in human obesity. centrating hormone (MCH), not to be confused
Leptin decreases when adipose stores decrease, with the MSH, stimulates food intake.
and low levels appear to act as a signal to the Monogenetic loss-of-function mutations in
organism to increase caloric intake. each of these steps have been described in rare
Etiology of Obesity 331

individuals. A more common single-gene muta- these drugs (a combination of fenfluramine with
tion due to early-onset obesity is a loss-of-­function phentermine was called fen-phen). Sibutramine,
mutation of the MC4R gene which is found in a serotonin and norepinephrine reuptake inhibi-
2–5 % of the heaviest individuals in several dif- tor, has been withdrawn by the FDA due to con-
ferent geographic locations. Patients have pro- cern over cardiac side effects.
gressive obesity, hyperinsulinism and insulin Endocannabinoids (eCBs) are derivatives of
resistance, increased lean mass, increased growth, arachidonic acid (AA) and resemble prostaglan-
and advanced bone age. Remarkably, some vari- dins in structure; these molecules play a role in
ants of the MC4R gene are protective of develop- reproduction, growth and development, and learn-
ing obesity. ing. The endocannabinoid system not only affects
Mutations affecting the cleaving of POMC emotions but also regulates appetite. The eCBs also
lead to early-onset and progressive obesity and affect energy homeostasis and eating behavior by
reddish hair due to a lack of MSH and secondary increasing the palatability of sweet and fatty tastes.
adrenal insufficiency due to the lack of ACTH. eCBs are elevated during fasting and decrease with
Prohormone convertase-1 (PC-1) deficiency is a satiety. Antagonists of the eCB system are being
cause of hypogonadotropic hypogonadism, but investigated for treatment of obesity.
since a mutation affects other preprohormones, The vagus nerve senses the fullness of the
patients are also likely to demonstrate ACTH stomach and duodenum and acts as a satiety sig-
deficiency, elevated levels of proinsulin, GI dis- nal to the central nervous system.
tress, and obesity. The WAGR syndrome Thus a simplified account of the CNS control
(#194072 ICD + WILMS TUMOR, ANIRIDIA, of appetite includes the following steps: (a)
GENITOURINARY ANOMALIES, AND Leptin stimulates production of POMC, (b)
MENTAL RETARDATION SYNDROME; which can be cleaved by enzyme action to MSH,
WAGR) combines numerous congenital defects which in turn (c) interacts with melanocortin 4
and in some patients, obesity related to decreased receptors (MCR4) to reduce food intake; (d) neu-
brain-derived neurotrophic factor; thus patients ropeptide Y (NPY) is a potent orexigenic (agent
may have a combination of aniridia, hypospadias, to increase feeding), and (e) AGRP, produced in
cryptorchidism, uterine malformations, nephro- the same neuron as NPY, competes with MSH for
blastoma (Wilms tumor), renal failure, and devel- the MCR4 and thereby also increases feeding.
opmental delay. Damage to one area of the hypothalamus may
Low dopamine levels, involved in pleasure affect these pathways to increase appetite, or
sensations, increase appetite, and this pathway is alternatively, damage to other locations of the
targeted for the development of medications for CNS will decrease appetite.
the treatment of obesity which will raise dopami-
nergic effects. Norepinephrine (NE) and sero-
tonin exert suppressive effects on appetite, Dietary Intake
which led to the development of medications that
amplify or mimic the action of these two neu- The treatment of obesity in adults brought about
rotransmitters. Selective serotonin reuptake the development of an exceptional number of
inhibitors used in psychiatry have a similar effect. specialty diets. Liquid diets, restrictive diets that
Phentermine is an NE reuptake inhibitor that is eliminate certain nutrients, and truly bizarre
still in use, but phenylpropanolamine was with- approaches are provided in the multibillion-dollar
drawn from the market when it was implicated in diet book industry. In spite of proliferation of
hypertension, stroke, and myocardial infarction. such plans, the general consensus of available
The widespread use of fenfluramine, a 5-HT research is that calories are responsible for the
uptake inhibitor, and of dexfenfluramine was dietary component of obesity, and it is calories
halted because of the increased risk of cardiac that should be limited to prevent the development
valve abnormalities that accompanied the use of of obesity or to treat, as long as restriction is not
332 13 Obesity

so severe as to interfere with normal growth. Thus elimination of juice from the diet is helpful,
Even a healthy diet can contain too many calories but this suggestion surprises some parents.
due to second and third servings. The clinician Water is the appropriate beverage of older
should approach the family whenever possible children as long as there is appropriate intake of
with the help of a dietitian by analyzing what milk to allow calcium and vitamin D intake.
they are eating and finding what they might Portion sizes have grown in the USA over the
accept as changes which would benefit the child. last decades and so have the plate sizes. It is
The clinician may have less experience with eth- important to evaluate the size of portions pro-
nic diets. Nonetheless every ethnic diet, while vided to a child and emphasize that eating sec-
sometimes having a tendency toward increased onds and thirds are certainly not healthy habits
calories, often can be manipulated to increase although many children demand these extra serv-
consumption of lower caloric foods. Asian diets ings. If the child remains hungry after an appro-
may have excessive rice which increases caloric priate serving, low- or no-calorie snacks can be
intake, but there are also vegetables which can be offered such as carrots, celery, or low-calorie
stir-fried or, even better, steamed to be part of a cheese, but a whole new dinner plate full of food
healthy diet. Spanish diets may contain foods is not appropriate. The MyPlate model will assist
with high carbohydrate content or heavily fried in determining appropriate portion sizes of meal
foods but also contain vegetables and fruit (even components (Fig. 13.3).
cactus) which can be introduced to the diet in a There is no question that a change in diet in a
healthy manner. The complexities of these issues child can lead to conflict in some homes which
point out why the well-trained dietitians familiar must be avoided. At times, one parent may be on
with pediatrics become so important. board with an appropriate change in diet only to
Simple approaches to modify diet can be pro- find that the other refuses to give up his or her
vided however. Excessive sugar intake is acknowl- soft drinks or chips or other highly caloric foods
edged as a major threat to health and can be that they think they deserve after working a hard
reduced as a first step. The World Health day. This presents quite a problem with no easy
Organization recommends decreasing total calo- answer except to suggest that the offending food
ries from added sugars to less than 5 %. The substances need to be kept somewhere else
American Heart Association recommends that no other than the house in which the child lives so
more than half of a person’s discretionary calorie that the stores of these foods will not be available.
allowance (daily “empty calorie” allowance) A problem that is virtually impossible to address
should be spent on added sugars. Any type of is the divorced family when one parent is able to
sugar-sweetened beverage should be eliminated control calories and then the other parent cares
from the child’s diet immediately. High-fructose for the child for the weekend and treats him or
corn syrup contains a mixture of glucose and fruc- her to anything he or she wants. Grandparents
tose similar to the composition of table sugar must participate in the approach to weight loss if
although the formulation does change between they care for the children. Children may go to
beverages and between companies. High-fructose their grandparents’ houses before school, and
corn syrup is now used in a wide variety of foods. some may ingest two breakfasts before school
But the endocrine system does not respond to starts: one breakfast at the grandparents’ house
fructose as it responds to glucose. Fructose inges- and another breakfast at school. It is clear that the
tion does not cause elevated insulin levels or leptin food environment is a complex issue and innova-
levels as does glucose, and some authorities feel tive solutions are required.
that fructose is a bona fide toxin to many meta- It is virtually impossible to “exercise off a bad
bolic processes. All fructose found in fruit comes diet.” One children’s meal at a popular fast-food
in a package combined with healthy vitamins and restaurant might contain 750 calories (some con-
fiber, but when the fruit is mashed into pulp, the tain over 1,400 calories). The amount of activity
liquid becomes a beverage with a very high calo- necessary to exercise off that single child’s meal
rie content equal to sugar-sweetened beverages. will exceed 3 h on an exercise bicycle, an
Etiology of Obesity 333

Fig. 13.3 (a) The MyPlate plate model developed by the priate portions as a wall chart. (c) The MyPlate model in
United States Department of Agriculture as a placemat. Spanish. Courtesy of Marilyn Townsend UC Davis with
(b) The MyPlate model with examples of food and appro- permission

e­ xpenditure that most providers are unlikely to mote increased fat deposition, while a low glyce-
accomplish and certainly most children will not. mic index meal such as beans or legumes will
Eating out frequently is a reliable source of produce a lower peak of glucose and therefore a
excess calories; in restaurants, the cost of the lower peak of insulin secretion. In some cases,
labor is higher than the cost of the food, so man- the preparation of a food such as potatoes will
agers give larger servings to try to satisfy the cus- influence the glycemic index complicating the
tomer that the meal is worth the cost. Eating out development of a low glycemic index diet; the
usually involves increased salt intake, and pro- glycemic index of a baked potato is 85 and of a
cessed foods are common. It is best to counsel the boiled potato is 50. It is evidenced that the low
family to eat at home and to follow healthy habits glycemic index leads to less weight gain in chil-
they have learned in the clinic. dren than a high glycemic index meal. The most
The type of carbohydrate ingested has effects recent studies confirm that control of calories is
on the rise in glucose and insulin in the postpran- paramount regardless of glycemic index or load.
dial state, and these characteristics have been Indeed, the diet one can accept and follow is the
used to advantage in the development of a one that will exert effects.
healthy diet. Glycemic index refers to the area There is much benefit from increasing fiber in
under the curve of glucose values by time after the diet of children and adults as present intake is
ingesting a meal of carbohydrates. A high glyce- considerably lower than previous eras. A high-­
mic index meal with carbohydrates that can be fiber diet leads to a lower rise in glucose in the
quickly broken down such as bread and rice leads postprandial state and thereby a lower rise in insu-
to a rapid rise in glucose which will trigger a lin and a lower degree of lipogenesis. A high-­fiber
rapid and elevated rise in insulin which may pro- diet will slow absorption of triglycerides and
334 13 Obesity

Fig. 13.3 (continued)
Etiology of Obesity 335

Fig. 13.3 (continued)
336 13 Obesity

improve fermentation of triglycerides in the lower function. The microbiome in turn is influenced by
gut. In sum, the effect of a high-fiber diet is to dietary composition and even exercise. Probiotics
increase insulin sensitivity that will thereby tend to are the beneficial bacteria which are found in fer-
improve lipid profiles. Fiber also will tend to pro- mented foods like yogurt, kefir, cheese, sauer-
mote satiety and tend to decrease caloric intake. kraut, kimchee, and kombucha, among others.
Prebiotics which provide fuel for gut microbes are
the nondigestible carbohydrates found in plant
Energy Expenditure foods including fruits, vegetables, whole grains,
legumes, nuts, and seeds, among others.
An increase in fat tissue occurs because of There is evidence that maternal factors during
increased caloric intake (by ingestion of food and pregnancy likewise influence the development of
drink) or decreased total energy expenditure (TEE). the infant’s microbiome. There is a difference in
TEE consists of the sum of (a) basal metabolic rate the intestinal flora between children born by vag-
(BMR) (resting energy expenditure (REE) is simi- inal delivery and those by C-section. Breast milk
lar to BMR but is measured in the postabsorptive contains prebiotics and also probiotics which
state, while basal metabolic rate is measured after introduce and support the beneficial bifidobacte-
fasting), (b) the thermic effect of feeding, and (c) ria in the infant’s gut.
nonresting energy expenditure. Nonresting energy There are correlational studies in human
expenditure is composed of exercise and non-exer- beings showing a difference in the microbiome
cise activity thermogenesis (NEAT, e.g., fidgeting). in obese and lean children and adults. This,
No consistent pattern of decreased basal metabolic added to experimental studies in animal models
rate accounts for the difference between children that provide cause-and-effect data, suggests that
with obesity and those who are not obese in most, the microbiome may have real importance in the
but not all, situations. African-American girls obesity epidemic and that manipulation of the
appear to have a lower basal energy expenditure microbiome with probiotics might be a future
than that of girls of other ethnic groups, however. therapy for obesity.
The balance of caloric intake and expenditure Although not related to gut flora, neutralizing
is remarkably sensitive. An increase of only antibodies to adenovirus 36 are found in increased
36 kcal of intake per day, equivalent to a “pat” of prevalence in obese adults and children although
butter, a change in dietary intake below the limit no cause-and-effect relationship to adenovirus
of detection in most clinical studies, leads to infection is proven in human beings.
13,140 calories per year, while 1 lb of fat equals
3,500 calories of intake; all things being equal, the
metabolic processes vary so much that this equiv- Epigenetics
alency is not strictly accurate but can be used for
illustrative purposes. Obesity is often the result of There is increasing evidence that epigenetics
a subtle but long-term increase in caloric intake. affects the development of obesity in the child.
While genetic mutations involve changes in the
base sequence of DNA, epigenetics is a revers-
Gut Microbiome ible process that does not change the base pairs,
but instead, reversibility chemically modifies
There are estimates that an individual has 10–100 DNA or the associated histones. This may occur
times as many microbes in their gut as there are by methylation of DNA (addition of methyl
cells in the body and that the microbiome contains groups to DNA) or posttranslational modification
100 or 1,000 times as many genes as there are of histone proteins by methylation or acetylation.
genes in our human genome. The gut microbiome These changes in DNA can progress through
influences food utilization and regulates aspects mitotic cell division and can determine whether a
of the intestinal barrier and intestinal immune gene is expressed or not expressed at a given time
Etiology of Obesity 337

development. Epigenetic modification appears to environmental factors. The genetic makeup of the
explain the phenomenon of the early origins of human population cannot have changed over the
adult disease often referred to as the Barker last 30 years sufficiently to cause the obesity epi-
hypothesis. Thus epigenetic modifications of the demic. Thus environmental factors are considered
fetus by the metabolic milieu of the mother lead a major influence on obesity and are targeted as
to changes in the metabolism of her child. In fact, modifiable influences. Environmental factors
the modification in an ovum in the individual’s account for changes in diet and activity which are
grandmother (where all the mother’s ova are the proximate factors that lead to increased weight.
formed) by changes in the grandmother’s endo- The environment of the home exerts a pro-
crine milieu, according to increasing evidence, found effect. Television watching is an empiri-
can exert effects expressed in the grandchild. cally proven factor in the development of obesity.
Thus one- or two-generation effects can occur The more an individual watches television as an
from, for example, diabetes or obesity in a preg- adult or child, the higher likelihood of develop-
nant woman. Epigenetic effects are felt to be ing obesity. The reasons are not clear, but the
responsible for aspects of obesity, type 2 diabe- thousands of calories of fast food advertised per
tes, and cardiovascular disease, just to name a hour during children’s television programs and
few conditions based in the early environment. the lack of physical activity during the time that
Epigenetic effects can occur because of overnu- the individual watches television are likely some
trition, undernutrition, or toxin exposure. There of the influences. Television sets are now found
is a U-shaped curve in which smaller-for-­ in an increasing percentage of children’s bed-
gestational-­ age infants (not necessarily classic rooms, and their very presence is associated with
SGA weights) or heavier infants (not necessarily increased obesity rates. Decreasing television
extending into the macrosomic range) develop viewing has been linked in experimental studies
insulin resistance and a tendency toward type 2 to maintaining a more stable BMI during child-
diabetes and obesity later in childhood and in hood. There are other screens than television
adulthood. However, the rate of growth in the screens in the child’s life, and there is evidence
postnatal period also appears to cause epigenetic that time spent viewing video games and comput-
modifications in metabolism. Maternal weight ers is likewise linked to the development of obe-
gain also appears to influence the risk of develop- sity. Some electronic games require physical
ment of type 2 diabetes and metabolic syndrome activity but have not been shown to be beneficial
later in life. Animal data is stronger than human in the treatment of obesity in all studies.
data, but the diet of the mother, in terms of the Electronic screens also affect sleep. It is well
components of carbohydrate and lipid composi- established that obesity can be linked to obstruc-
tion, may also affect metabolic programming of tive sleep apnea (OSA) which will decrease
the fetus. On the positive side, breast-feeding a effective sleep (see below). But limitation or vari-
macrosomic infant has been shown to lower the ation in sleep time can also change carbohydrate
risk for diabetes. While epigenetic modification metabolism, increasing insulin resistance.
can be shown to cause biochemical modifications Children living in disordered households and
of DNA, some of the factors thought to be respon- noisy neighborhoods as well as those with elec-
sible for epigenetic mechanisms may also be fac- tronic screens in their rooms may be the ones
tors which influence gut biome development. with an increased tendency toward obesity due to
this factor. Reading on e-readers or surfing the
Web before bed disturbs sleep due to the tone of
The Obesogenic Environment the light, while reading paper books does not
have the same effect before going to sleep.
Obesity develops in an individual with a suscepti- The microwave oven in the home has allowed
ble genetic makeup who is exposed to an “obeso- more children to be responsible for much of
genic” environment, a term that includes a host of their diet, and the foods that are characteristi-
338 13 Obesity

cally microwaved tend to be highly caloric, Endocrine Causes


often highly salted processed foods. “Latchkey”
children are those coming home from school to Many parents blame imbalance of “hormones”
an empty house while their parents are working or common conditions like hypothyroidism for
one or more jobs. At home may be a ready sup- obesity in their children and wish for an easy
ply of highly caloric foods and the television set therapy for these conditions. In truth, while in
which when combined increase likelihood of exceptionally rare circumstances hypothyroid-
obesity. ism can cause weight gain which is treatable
The environment of the child extends further with thyroid hormone, this is not the cause of the
than the home in which they live. Many disadvan- obesity epidemic and not the treatment for the
taged youth live in communities in which it is vast majority of children. There are endocrine
either truly dangerous to play outside or where conditions which can lead to an obese appear-
parents perceive there is a danger which leads the ance, and they cluster around problems of
parents to keep the child inside rather than allow- growth in most cases. Pseudohypoparathyroidism
ing outdoor play. Zip codes with the lowest with Albright’s hereditary osteodystrophy and
income frequently lack playground, parks, and Cushing syndrome combine short stature with a
access to indoor gymnasiums as well. Schools are chubby or overweight appearance. Most children
often built on the cheapest real estate which often with standard (sometimes called “exogenous”)
are located miles from where children live, neces- obesity are taller than average, eliminating
sitating a car or bus ride rather than a walk to almost all endocrine causes. Obese individuals
school, cutting down energy expenditure on with taller stature have low response of GH
school days. There is also a tendency to build secretion after stimulation but nonetheless have
schools without gymnasiums, replacing them advancement of bone age and normal IGF-1 val-
with multipurpose rooms which by definition are ues, while bona fide growth hormone deficiency
used for various activities, some of which likely presents with short stature, delayed bone age,
cut down physical activity. Likewise in some and a secondarily increased BMI due to the lack
states, gym teachers must have special licenses, of normal lipolytic effect of GH. Hyperinsulinism
eliminating volunteer teachers of other subjects could lead to tall stature but will be accompanied
from leading activities that would increase the by such severe hypoglycemia that it is not likely
child’s activity. Low-income neighborhoods fre- to be missed. In Chaps. 5 and 9, Prader–Willi and
quently lack grocery stores, and even if there are Bardet–Biedl syndromes are described which
small stores, they are often convenience stores combine short stature and obesity as well. It is
without fresh fruits and vegetables (food deserts). notable that ghrelin levels are higher in Prader–­
Suburban neighborhoods may lack sidewalks to Willi syndrome than in children with similar val-
allow safely walking to stores and schools even ues of BMI who do not have the syndrome.
when the distance makes it feasible. The obesity Suffice it to say that obesity can precipitate endo-
epidemic in a very substantial manner is rooted in crine disorders, but endocrine disorders did not
socioeconomic problems and poor city planning cause the obesity epidemic, and endocrine treat-
of the built environment. ments will not solve this problem.
There is increasing evidence that endocrine Sophisticated parents will always request that
disrupters exert significant effects on the develop- testing be performed for hypothyroidism, and a
ing fetus and child. Cell culture and animal exper- provider may consider carrying out such tests,
iments implicate certain substances, but causal but they are going to be extremely low yield.
effects in human beings cannot be proven yet. Children’s serum TSH values tend to be higher
There are correlations between urine excretion or than adults, and children with obesity also tend to
plasma levels of certain endocrine disruptors such have higher TSH values, but these do not indicate
as bisphenol A (BPA) and the development of hypothyroidism in the vast majority of individu-
obesity, but correlation is not yet causal. als (see Chap. 6).
Comorbidities of Childhood and Adolescent Obesity 339

Hypothalamic obesity is seen in some individu- the development of obesity, but there is no ques-
als with damage to the ventral medial nucleus of tion that obesity is an insulin resistance condition.
the central nervous system. It is most well known
following surgery for craniopharyngioma. The sit- Antibiotics
uation in which even with food restriction weight Recent evidence reports an association of expo-
may increase as the brain is incapable of respond- sure to broad-spectrum antibiotics in the first 2
ing to leptin signals and treatment of the patient’s years of life with an increase in BMI, especially in
condition with virtual starvation is counterproduc- boys. Narrow-­spectrum antibiotics did not seem to
tive. Treatment with somatostatin agonists to sup- exert the same effect. In many cases, the antibiot-
press insulin secretion has been helpful in some ics are used for viral infections where they were
patients in clinical studies but is not FDA approved. not indicated, making such a relationship all the
more unfortunate. The etiology of this relationship
is not clear but may relate to a change in the gut
Iatrogenic Causes microbiome and overuse of antibiotics in children.
More research is needed to confirm this finding.
Antipsychotic Medication
It is well known that high doses of glucocorti- Central nervous system surgery
coids can cause Cushing syndrome. Perhaps less or injury
well known but clearly established is weight gain The hypothalamus controls appetite. Injury to the
with atypical (second generation) antipsychotic hypothalamus by a tumor, treatment for a tumor or
and other types of psychiatric medication; 80 % by other trauma can cause insatiable appetite
of children and teenagers taking atypical antipsy- known as hypothalamic obesity. Increased insulin
chotic medications gain weight often to a signifi- secretion is proposed as one cause for this situa-
cant degree leading to obesity. Examples of tion and suppression of insulin with, eg., octreo-
medication ­fostering weight gain are clozapine, tide has been used as a treatment modality.
olanzapine, quetiapine, divalproex, paroxetine,
doxepin, imipramine, nortriptyline, and mono-
amine oxidase inhibitors (MAOIs) such as Nardil
(phenelzine), Parnate (tranylcypromine), and  omorbidities of Childhood
C
Marplan (isocarboxazid) which cause an and Adolescent Obesity
increased risk for type 2 diabetes in treated chil-
dren as well as a higher risk if the child is a girl An excess of visceral fat that surrounds the
and if the treatment is given in the teenage years. abdominal organs and ectopic fat in muscle, liver,
The psychiatric literature recommends withhold- and other organs, rather than the more visible
ing such treatment if alternative methods of ther- subcutaneous adipose tissue, is related to many
apy are available and that if such treatment is of the metabolic derangements of obesity such as
used, dietary recommendations be given with the insulin resistance and hyperlipidemia. Waist cir-
medication and screening for comorbidities of cumference reflects the amount of visceral fat
obesity including type 2 diabetes be carried out. and is measured regularly in adults, and this will
likely become common practice in children as
Insulin well in the future as standards become more
Insulin treatment of diabetes can cause weight available although accurate repeatable measure-
gain; some teenage girls with diabetes skip insulin ments are difficult to achieve (standards are
doses to counteract this tendency, sometimes with found in Table 13.2). Increasing waist circumfer-
tragic consequences. A healthy balanced diet with ence is related to insulin resistance, blood pres-
reasonable carbohydrate intake requiring reason- sure, lipid levels, and metabolic syndrome.
able insulin dosing is the best approach to avoid About 30 % of adult obesity has its origin in
this situation. There is considerable controversy childhood obesity. With the more severe and the
over the effect of elevated endogenous insulin on younger onset of obesity in a child and the
340 13 Obesity

p­ resence of obesity in one or more of parents, the Weightism is prevalent in society, and chil-
likelihood of the child with obesity becoming an dren may be the brunt of it as young children are
obese adult increases. Tracking of obesity from already the target of teasing and ridicule when
childhood into adulthood becomes more certain overweight. Young children may even express
as the child becomes older and comes closer to the aspects of weightism as children younger than 10
adult age group. Further, an adult who was obese years demonstrate preference for playmates with
as a child has substantially more severe comor- a thinner body habitus. The economic well-being,
bidities of the obesity during the adult years. BP is likelihood of advanced education, and likelihood
higher, and lipid profiles are more adverse in of marriage are decreased with obesity in some
adults who were overweight or obese as children. studies.
For example, the Indian Health Service data
shows that the highest BMI values in childhood
lead to greater risk of early death in the adult. Type 2 Diabetes

Type 2 diabetes mellitus was rarely seen in child-


Psychological Conditions hood and is rising in childhood but cannot be
considered as epidemic as it affects 1.7 per 1,000
While there have been significant numbers of children lessons. American Indians have a 2–40
studies on the psychological effects of obesity in times increase in prevalence with other ethnic
childhood, there are concerns that the samples groups also having higher percentages compared
might not be representative of the population that to Caucasians. Diagnosis of type 2 diabetes mel-
does not seek help or did not come to the clinic. litus rests on particular blood sugar values (see
Nonetheless, obesity has been shown to decrease Chap. 11) and not on increased fasting insulin
quality of life and have a relationship to the concentrations (see Chap. 13).
development of depression. Anorexia nervosa is
well known to be a weight-losing condition, but a
majority affected in some studies previously suf-  he Metabolic Syndrome and Insulin
T
fered from obesity. Likewise in the natural h­ istory Resistance
of the condition, in an obese person who later
develops anorexia nervosa, symptoms of the con- The obesity epidemic has caused pediatricians to
dition preceded the loss of weight, and the clini- shift focus to diagnosis and treatment of many
cian must have a high index of suspicion for conditions previously found only in adults. The
disordered eating behavior. It goes without say- metabolic syndrome was previously found in
ing that the clinician treating a child with over- middle-aged adults, often in males, but now is
weight or obesity must have extreme caution and found in teenagers and even prepubertal children
not adversely affect their psychological state: with increasing frequency. The diagnosis of the
sensitive and careful approaches to the child with metabolic syndrome in adults does not directly
obesity have not been linked to the development transfer to the metabolic syndrome in children
of anorexia nervosa or other eating disorders. and adolescents and numerous publications have
Individuals with preexisting psychological different definitions of the metabolic syndrome
conditions may be predisposed to developing
­ in adolescence. A recent consensus conference
obesity which might be particularly difficult to suggested coordinating approaches to the meta-
treat. There are screening instruments for depres- bolic syndrome in adolescence so that studies can
sion available. The Pediatric Symptom Checklist be compared in the future and that studies should
is in the public domain (http://www.massgeneral. use the same definition. The International
org/psychiatry/services/psc_home.aspx) while Diabetes Federation suggested a new definition
others are availavle for purchase. Children with of the metabolic syndrome at 10 years of age; for
developmental delay are frequently seen with over 16 years of age, an adult definition can be
obesity or overweight. used. For children between 10 and <16 years of
Comorbidities of Childhood and Adolescent Obesity 341

Table 13.1  Estimated value of waist circumference for percentile regression in nationally representative samples of
American children according to sex
Percentile for boys Percentile for girls
10th 25th 50th 75th 90th 10th 25th 50th 75th 90th
Intercept 39.7 41.3 43.0 43.6 44.0 40.7 41.7 43.2 44.7 46.1
Slope  1.7  1.9  2.0  2.6 3.4  1.6  1.7  2.0  2.4 3.1
Age (years)
2 43.2 45.0 47.1 48.8 50.8 43.8 45.0 47.1 49.5 52.2
3 44.9 46.9 49.1 51.3 54.2 45.4 46.7 49.1 51.9 55.3
4 46.6 48.7 51.1 53.9 57.6 46.9 48.4 51.1 54.3 58.3
5 48.4 50.6 53.2 56.4 61.0 48.5 50.1 53.0 56.7 61.4
6 50.1 52.4 55.2 59.0 64.4 50.1 51.8 55.0 59.1 64.4
7 51.8 54.3 57.2 61.5 67.8 51.6 53.5 56.9 61.5 67.5
8 53.5 56.1 59.3 64.1 71.2 53.2 55.2 58.9 63.9 70.5
9 55.3 58.0 61.3 66.6 74.6 54.8 56.9 60.8 66.3 73.6
10 57.0 59.8 63.3 69.2 78.0 56.3 58.6 62.8 68.7 76.6
11 58.7 61.7 65.4 71.7 81.4 57.9 60.3 64.8 71.1 79.7
12 60.5 63.5 67.4 74.3 84.8 59.5 62.0 66.7 73.5 82.7
13 62.2 65.4 69.5 76.8 88.2 61.0 63.7 68.7 75.9 85.8
14 63.9 67.2 71.5 79.4 91.6 62.6 65.4 70.6 78.3 88.8
15 65.6 69.1 73.5 81.9 95.0 64.2 67.1 72.6 80.7 91.9
16 67.4 70.9 75.6 84.5 98.4 65.7 68.8 74.6 83.1 94.9
17 69.1 72.8 77.6 87.0 101.8 67.3 70.5 76.5 85.5 98.0
18 70.8 74.6 79.6 89.6 105.2 68.9 72.2 78.5 87.9 101.0
Data from Fernández JR, Redden DT, Pietrobelli A, Allison DB. Waist circumference percentiles in nationally
representative samples of African-American, European-American, and Mexican-American children and adoles-
cents. J Pediatr. 2004 Oct;145(4):439–44, with permission

age, the metabolic syndrome is diagnosed by a The basis of the metabolic syndrome is insulin
waist ­circumference ≥90th percentile or adult resistance usually caused by obesity and predom-
cutoff if lower, triglycerides ≥150 mg/dL, high- inantly visceral adiposity. Thus increased BMI
density lipoproteins (HDL) <40 mg/dL, elevated and increased waist circumference are suggested
blood pressure over the 95th percentile for gen- as basis for the diagnosis. Fasting lipid studies
der age and height (use standards presented in demonstrate low HDL cholesterol and elevated
Table 13.3 or systolic ≥130/diastolic ≥85 mmHg), triglycerides. Glucose intolerance and type 2 dia-
and fasting blood sugar ≥100 mg/dL or known betes are frequently manifestations of the meta-
type 2 diabetes. bolic syndrome. While the basis of the metabolic
Waist circumference percentile is part of the syndrome appears to involve insulin resistance,
diagnosis, but charts for waist circumference are single measurements of basal insulin in a child
not readily available; they are provided in this are of little utility in the diagnosis of the meta-
publication as Table 13.1. Thus, a clinician might bolic syndrome. The gold standard evaluation for
use a high BMI for a child or teenager, over the the metabolic syndrome is the euglycemic clamp
95th percentile, hypertension, elevated triglycerides which is beyond the scope of any clinical office
with low HDL cholesterol, and prediabetes or true encounter. Insulin sensitivity decreases normally
type 2 diabetes for a diagnosis of metabolic syn- during puberty, and the metabolic syndrome
drome. No definition for the metabolic syndrome is often becomes manifest during this age range.
provided for children under 6 years of age, but the Elevated basal insulin levels are frequently found
astute clinician will note who seems to be heading in obesity because obesity is an insulin-resistant
in the direction of the metabolic syndrome and state. However, elevated ­ insulin levels do not
institute anticipatory guidance for the parents. indicate that the child has or will get type 2 dia-
342 13 Obesity

betes, just that there is insulin resistance, which is the inflammatory state will be in elevation of
expected in obesity. Thus the measurement of C-reactive protein (CRP) measured in sensitive
insulin is not usually used in the definition of the assays; IL6 rises as well in obesity in the meta-
metabolic syndrome. bolic syndrome. Cytokine levels are not part of
Insulin resistance in obesity and metabolic the definition of the metabolic syndrome, and
syndrome appears to be selective as some organs analyses are not carried out in most clinical situ-
are more resistant to insulin than others. The ations, but it is important to consider inflamma-
liver is resistant to the effects of insulin on tion as a component of the metabolic syndrome
­carbohydrate metabolism such as the inhibition in childhood to explain why it can be so damag-
of gluconeogenesis which leads to glucose intol- ing to cardiovascular health later in life. Further,
erance. Further, the characteristic insulin effect obesity is a state of elevated reactive oxygen
of inhibition of lipolysis fails, leading to elevated species which may arise in part from the effects
circulating triglycerides. The metabolic of the inflammatory cytokines. Reactive oxygen
­syndrome is characterized by ectopic fat deposi- species cause damage to DNA and cellular func-
tion, and fat may be found in the liver, pancreas, tion. The sum of these activities is long-lasting
omentum, myocytes, and visceral organs, and changes in health.
ectopic fat deposition is related to dyslipidemia.
The ratio of visceral to subcutaneous fat is
directly related to insulin resistance. Polycystic Ovarian Syndrome
Insulin resistance normally increases at
puberty, and this is likely the reason that T2DM Polycystic ovarian syndrome or PCOS is the
appears usually after puberty. Further, Caucasians most common reproductive problem of women
are normally more insulin sensitive than African and often starts in the adolescent years. It is char-
Americans, Hispanic Americans, Asians, and acterized by irregular or absent menses and
American Indians, and this is why the latter increased prevalence of acne and hirsutism and
groups have the highest prevalence of T2DM. can often be found in a familial pattern. Patients
Increased cortisol can foster visceral are not always obese, but when they are, the syn-
adiposity, and obese individuals have decreased drome is worse. PCOS presents aspects of the
11-β-hydroxysteroid dehydrogenase-1 (11b2HSD1) metabolic syndrome in women. A more complete
which converts the active moiety cortisol to the discussion is found in Chap. 9.
inactive-form cortisone. Thus the increased
­cortisol effect may promote the metabolic syn-
drome. While stress in adults which leads to  yperlipidemia and Risk
H
increased cortisol secretion is a factor to also pro- for Cardiovascular Disease
mote the metabolic syndrome, this association is
not established in children. Dyslipidemia is manifested as elevated low-­
The metabolic syndrome is accompanied by density lipoprotein (LDL) cholesterol, non-HDL
high levels of insulin. Even though the body is cholesterol, and triglycerides but decreased HDL
resistant to insulin in terms of carbohydrate cholesterol when measured after a 14-h fast
metabolism which leads to glucose intolerance, (except for water). Dyslipidemia is a risk factor
elevated insulin can produce many other comor- for cardiovascular disease, and the prevalence of
bidities. For example, elevated insulin levels can dyslipidemia is rising with the obesity epidemic.
interact with IGF-1 receptors to stimulate growth There has been previous evidence of the presence
of cells, increasing the risk of various types of of atherosclerosis in young adults dating from
cancers later in life. the Korean War and more recently in children
The metabolic syndrome is a condition of and teenagers from the Bogalusa heart study.
increased inflammation as is obesity itself. Thus Atherosclerosis is now clearly established in
inflammatory cytokines are elevated, but in the childhood and adolescence even though throm-
clinic, the most easily measurable indication of bosis does not usually occur until adulthood.
Comorbidities of Childhood and Adolescent Obesity 343

A brief summary of lipid metabolism involves taken up by LDL receptors on the liver mediated
the following. Lipids are energy-rich com- by apoprotein E (apo E). Very-low-density lipo-
pounds; triglycerides provide energy storage in protein (VLDL) is produced in the liver by com-
adipocytes and muscle cells, while cholesterol bining cholesterol and triglyceride produced
serves many roles in cell membranes, steroids, endogenously, rather than ingested, with apoB-­
bile acids, and signaling molecules. Because lip- 100(apoB), and the VLDL is secreted into the
ids are insoluble since they are hydrophobic, lymph and also transferred into the bloodstream
they are transported in the blood with hydro- where much is converted to LDL. VLDL
philic lipoproteins, apoproteins, or apolipopro- increases with intrahepatic FFA as occurs with
teins. The surface proteins of the spherical high-fat diets, obesity, and diabetes. The VLDL
lipoproteins serve as ligands and cofactors for surface contains Apo C-II which activates lipo-
the lipid-­processing enzymes. The size of the protein lipase on endothelial cells to break down
lipoproteins and their density, the ratio of lipid to triglycerides into FFA and glycerol. VLDL
protein, are the identifying characteristics that metabolism produces intermediate-density lipo-
define them. LDL cholesterol when elevated is a proteins (IDL) which are cholesterol-rich VLDL
risk factor for heart disease. The small dense and chylomicron remnants which can be metab-
LDL contain the most cholesterol esters and are olized by hepatic lipase to produce LDL. IDL
especially atherogenic; these molecules are contains Apo B.
associated with hypertriglyceridemia and insulin LDL cholesterol is the product of VLDL and
resistance. Further, if HDL cholesterol levels are IDL metabolism and contains the most cholesterol
decreased, the risk for atherosclerotic heart dis- of these molecules; LDL is associated with Apo B.
ease rises. Lipoprotein(a) (LP(a)) consists of an LDL mol-
Ingested dietary fat, mainly triglycerides but ecule and apoprotein A. This cholesterol-contain-
also cholesterol esters and phospholipids, free ing molecule is related to atherosclerosis, coronary
fatty acids (FFAs), among other lipids, is artery disease, and stroke, and LP(a) and LDL are
digested to fatty acids, monoglycerides, and the most atherogenic lipoprotein molecules.
unesterified cholesterol in the gut. These are LDL is taken up by LDL receptors on the
made soluble by bile acid micelles and are liver, and other tissues, mediated by Apo B and
absorbed through passive and active processes hepatic LDL receptors. There is a limit as to how
through the intestinal cellular wall. Once into the much LDL can be taken up, and the excess will
enterocytes of the GI tract, production of lipo- be oxidized which can lead to damage of epithe-
proteins within the intestinal cells proceeds by lial cells and contribute to atherosclerosis. High
attachment of apolipoprotein B-48 to a central LDL levels decrease LDL uptake, and lower LDL
core of nonpolar cholesterol esters and triglycer- levels increase the uptake of LDL.
ide and an outer region of free cholesterol and HDL brings lipids from the periphery to the
phospholipids (polar lipids) which constitutes a liver, and this process can remove lipids from
chylomicron. Thus the production of chylomi- atherosclerotic sites; HDL is antiatherogenic.
crons allows transport of the ingested triglycer- Apolipoprotein A is the main protein component
ides and cholesterol into lymphatic ducts where of HDL cholesterol.
they are secreted and transferred to the venous Endogenous cholesterol is synthesized by the
system. With the addition of other apolipopro- mevalonate pathway; the rate-limiting step is
teins to the chylomicrons, including apoC-II, HMG-CoA reductase (3-hydroxy-3-methyl-
C-III, and apoE (apoprotein E which were trans- glutaryl-­CoA reductase) which is located in the
ported by HDL), hydrolyzation of the triglycer- endoplasmic reticulum. HMG-CoA reductase is
ides within the apoC-II, C-III, and apoE proceeds inhibited by the class of drugs known as HMG-­
by lipoprotein lipase to produce fatty acids and CoA reductase inhibitors or statins which are
glycerol. In this process, lipoprotein lipase is ­frequently used for the treatment of hypercholes-
activated by apoC-II but inhibited by apoC- terolemia (statins are teratogenic). LDL attaching
III. The remaining smaller chylomicrons are to the LDL receptor in the liver leads to meta-
344 13 Obesity

bolic change, releasing cholesterol which inhibits percentile. Other conditions add further risk, but
HMG-CoA reductase. HMG-CoA reductase for the purposes of this discussion, the presence
inhibitors induce an increase in LDL receptors of diabetes of either type is the most important
which lowers plasma cholesterol, serving to risk factor (see Chap. 11).
decrease the risk of atherosclerosis. Cholesterol Universal lipid screening is recommended
esters are produced from free cholesterol by the between 9 and 11 and again between 17 and 21
action of lecithin–cholesterol acyl transferase. years unless there is a family history of dyslipid-
An increased ratio of apolipoprotein B (ApoB) emia at which time screening should be performed
to apolipoprotein A-I (ApoA1) is directly related at 2–8 years of age. Lipid screening is recom-
to LDL levels as ApoB is associated with LDL mended for BMI over the 95th %. Children with a
and ApoA with HDL. serious concern based upon family history should
While obesity tracks strongly in individual have fasting lipid screening done twice at least 2
between child and adulthood, meaning that child weeks apart but within 3 months with the average
with obesity has a higher likelihood of becoming a determining the future management approach.
obese adult, lipid abnormalities in childhood have Lipid panels are best determined in the fasting
a less robust tracking pattern, but it is still disad- state, and results are of concern if LDL choles-
vantageous to have dyslipidemia in childhood. terol >130 mg/dL, non-HDL cholesterol >145 mg/
The office visit as discussed in Chap. 2 is a dL, HDL cholesterol <40 mg/dL, and triglycer-
time to investigate a family history of cardiovas- ides >100 mg/dL if <10 years or >130 mg/dL if
cular events such as myocardial infarction or >10 years. If there is concern that the patient will
strokes. It is not adequate to simply ask if there is not come back to get a fasting lipid level, perform
anyone in the family with high blood pressure or a non-fasting lipid panel. For a non-fasting lipid
dyslipidemia but instead to ask if anyone is tak- level, the non-HDL cholesterol is calculated by
ing medications for high blood pressure or dys- the formula non-HDL cholesterol = total choles-
lipidemia because often, once successfully terol minus the HDL cholesterol. Non-HDL cho-
treated, the family thinks that a condition no lon- lesterol is abnormal if the non-HDL > 145 mg/dL
ger exists. Likewise, determination of early car- and the HDL < 40 mg/dL.
diovascular events or deaths even if unexplained As always, lifestyle change is the first
(early deaths are variously defined, but a useful approach to complications of obesity such as
definition is under the age of 55 in males or 65 in dyslipidemia. A heart-healthy diet and increased
females or younger) indicates a strong history of activity with a decrease in sedentary time are
cardiovascular risk. It is important to realize that always the basis of treatment. The amount and
the parents of our patients tend to be relatively types of fat in the diet are important in construct-
young and are still able to develop diabetes and ing a healthy lifestyle. Unsaturated fats are a
cardiovascular complications, so the family his- beneficial type of dietary fat and are usually
tory must be updated for each visit for each child. found in plants and include vegetable oils, nuts,
Risk factors should be evaluated in the indi- and seeds; unsaturated fats can be monounsatu-
vidual. Being born small for gestational age, rated or polyunsaturated, and omega-3 fatty
being an infant of diabetic mother, or having acids are a type of unsaturated fat. While there is
macrosomic birth weight all lead to increased some controversy as to the link between satu-
risk of insulin resistance and obesity later in life. rated fats and heart disease, saturated fats are
Moderate-level risk factors are considered to be characteristically thought to be adverse to health
hypertension not requiring drug therapy, BMI and should be replaced in the diet with unsatu-
equal to or >95th percentile and <97th percentile, rated fats but not replaced with foods containing
and HDL cholesterol <40 mg/dL. High-level risk refined carbohydrates. There is no controversy
factors are considered to be hypertension requir- that transfatty acids or transfats are adverse to
ing drug therapy (BP > 99th percentile + 5 mmHg), health; these hydrogenated vegetable oils which
current cigarette smoker, and BMI at the >97th become more solid with lower temperature are
Comorbidities of Childhood and Adolescent Obesity 345

linked to increasing LDL and decreasing HDL, Some parents find that there are cures on the
an inflammatory state, and insulin resistance. Internet for dyslipidemia such as red rice yeast
Until recently, most fast foods were laden with which contains agents similar to statins, but it must
transfatty acids, and stick margarine is another be emphasized to these parents that herbal
product of transfatty acids. Transfatty acids ­treatments are not regulated by the government and
should be eliminated from the diet as much as the side effects of this herbal cure are exactly the
possible. Transfatty acids are decreasing in the same as from the bona fide statin medications.
USA but may increase in the developing world. To emphasize again, statins are teratogenic, and
The effect of dietary cholesterol intake on the reproductive-aged females must be abstinent or on
lipid profile is not as strong as previously birth control while taking statin medication.
thought, but moderating cholesterol intake is still Niacin has been used in the past but recent
a recommended approach to diet. For our pur- studies demonstrate an increase in one unex-
poses, the recommended diet contains the follow- pected death per 200 niacin users in adult studies
ing: total fat 25–30 % of calories, saturated fat at which effectively dissuade use of niacin in chil-
<10 % of calories, and cholesterol intake <300 mg/ dren for dyslipidemia. There is little information
day. If the person’s LDL was further elevated, the on the use of fibrates and cholesterol-binding
diet is modified to contain saturated fat as 7 % of agents in children, and they should only be used
calories and dietary cholesterol no more than with a lipid specialist consultation.
200 mg/day. Plant sterols up to 20 g per day appear While lipid abnormalities develop with obe-
to be safe and effective in further lowering LDL sity, it is important to eliminate genetic causes of
cholesterol levels. If there is only an elevated tri- dyslipidemia or secondary dyslipidemia.
glyceride level, weight loss and a limited carbohy-
drate diet have been shown to be effective.  enetic Causes of Dyslipidemia:
G
Medications are limited to children over 10 Primary Hyperlipidemia
years of age who have met certain criteria. If the Familial hypercholesterolemia (#143890
total cholesterol is greater than 190 mg/dL and a I C D   +   H Y P E R C H O L E S T E RO L E M I A ,
child with no other risk factors fails to respond FAMILIAL) is an autosomal dominant condition
after 6 months of lifestyle modification or if the due to an abnormality of the LDL receptor and is
LDL cholesterol is between 160 and 190 with a the most common single-gene disorder leading
positive family history of risk factor in the to hypercholesterolemia. The prevalence of
­individual, medications may be considered. If the heterozygosity is 1,500 and homozygosity is
­
child has diabetes mellitus and the LDL concen- 1,000,000. Plasma cholesterol levels may rise to
tration is greater than 130 mg/dl (or even 100 mg/ 300–500 mg/dl in heterozygotes and as high as
dl according to some), medication might be con- 1,000 in homozygotes. Physical findings include
sidered if lifestyle modification failed. corneal arcus of the eye, xanthelasma of the skin,
The medications most often used in childhood tendinous xanthomas, and planar xanthomas in
dyslipidemia are HMG-CoA reductase inhibitors homozygotes.
which are commonly known as “statins.” Although Premature coronary artery disease may present
they are used in childhood with a generally safe in childhood in homozygotes or after age 30 years
profile, there are few long-term data and especially in heterozygotes. Dietary management is charac-
no data on outcomes many decades after childhood. teristically initiated in disorders of lipid metabo-
Knowing this, they still are instituted in appropriate lism, and medication may be considered for this
children. They are teratogenic so that any female of condition. National guidelines suggest implement-
reproductive age treated with this medication must ing a Cardiovascular Health Integrated Lifestyle
be on contraceptive agents or must be abstinent. Diet (CHILD-1) for the first 3–6 months, and if
Other side effects are elevation of hepatic enzyme results are not achieved, progression to the more
concentrations and elevated creatine kinase with lipid-­restrictive CHILD-2 diet (see ref #38 for
myopathy which may progress to rhabdomyolysis. these diet recommendations) is recommended.
346 13 Obesity

The addition of plant stanols to the diet has been achieve a BMI below the 85th percentile. Increased
demonstrated to lower cholesterol levels safely. activity is beneficial. A diet or additives containing
The goal of dietary management is to achieve an omega-3 fatty acids may decrease serum triglycer-
LDL cholesterol level less than or equal to 130 mg/ ide, but there has been recently suggestive links
dL. Over the age of 10 years, if LDL is not less between elevated levels of omega-3 fatty acids and
than 190 mg/dL or in the presence of a risk factor, prostatic carcinoma although an explanation for
less than 160 mg/dL, pharmacologic therapy, usu- this connection is not available. While statins can
ally with statins, can be initiated; sometimes this decrease serum triglycerides, fibrates are more
therapy is used in younger children with consulta- direct treatment for hypertriglyceridemia.
tion with a lipid specialist. Lipoprotein lipase deficiency (alleles of
Familial combined hyperlipidemia (#144250 +609708 LIPOPROTEIN LIPASE; LPL), some-
ICD+HYPERLIPIDEMIA, FAMILIAL COMBI- times known as familial chylomicronemia syn-
NED; FCHL) is an autosomal dominant condi- drome, is a group of rare conditions with various
tion with a prevalence of 1–2 % that is due to presentations and is generally due to a monoge-
overproduction of hepatic VLDL or LDL. While netic mutation in the lipoprotein lipase gene or
there is variation, the lipid profile characteristi- one of its cofactors. Thus lipoprotein cannot be
cally reveals elevated cholesterol, triglyceride, or metabolized, and chylomicrons and triglycerides
apoB found in a familial pattern. Xanthomas are accumulate, leading to a milky appearance to the
rare in this condition. Treatment by statins of the plasma in homozygotes. Manifestations occur in
hypercholesterolemia is considered. Treatment childhood, and along with very elevated triglyc-
of severely elevated triglycerides with fibrate eride levels, affected individuals also have
derivatives or fibrates increases degradation of abdominal pain, recurrent acute pancreatitis,
triglycerides and decreases production of VLDL eruptive cutaneous xanthomata, and hepato-
by the liver as well as raising HDL levels. Statins splenomegaly. Diagnosis is accomplished by
and fibrates are not to be used together because measurement of LPL enzyme activity after
they share a similar list of side effects including administration of heparin. Treatment is by dietary
elevated liver transaminase enzymes, myopathy, management to maintain triglyceride levels
and gastrointestinal symptoms. below 2,000 mg/dL to avoid GI pain and below
Familial hypertriglyceridemia (#145750 1,000 mg/dL ideally. Fibrates may be used if
I C D + H Y P E R T R I G LY C E R I D E M I A , dietary management is not successful.
FAMILIAL) is an autosomal dominant condition
with a prevalence of about 5–10 %. The cause of Secondary Hyperlipidemia
this condition is not definitely established, but it The prevalence of the hypertriglyceridemia is ris-
has been related to mutations in apolipoprotein ing in America, and it is associated with obesity
A5 gene (APOA5) and the lipase I gene (LIPI) on and insulin resistance which likewise cause
OMIM. The hypertriglyceridemia is caused by decreased HDL cholesterol values. Addressing
secretion of large VLDL particles laden with tri- the underlying obesity should improve the
glycerides. Atheroeruptive xanthoma is a feature hypertriglyceridemia.
of this condition. Triglyceride values are in the Combined hyperlipidemia can be found in
range of 200–500 mg/dL, VLDL values are ele- uncontrolled type 1 or type 2 diabetes mellitus
vated, and apoprotein C-II are low, while LDL and type 2 diabetes mellitus and is associated
and HDL cholesterol values are low to normal. with increased small dense LDL particles.
The condition is associated with an elevated risk Nephrotic syndrome and chronic kidney disease
for very premature atherosclerosis. will lead to dyslipidemia as will cholestatic liver
Dietary management of hypertriglyceridemia disease. LDL values are elevated in untreated
is suggested. The child is to follow a dietary pat- hypothyroidism. Cigarette smoking and alcohol
tern which involves decreasing sugar intake in use will likewise lead to dyslipidemia. Successful
solid or liquid foods and weight management to treatment of the underlying disorder or a change
Comorbidities of Childhood and Adolescent Obesity 347

in habits should resolve the secondary presenta- The latest national health and nutrition examina-
tion of combined hyperlipidemia. tion survey of the USA determined that 20 % of
Secondary hyperlipidemia can also cause children between 9 and 11 years already required
pancreatitis. an adult cuff, and 30 % of those children will
require a cuff larger than the standard adult cuff.
The appropriate cuff has an inflatable bladder
Hypertension width that is at least 40 % of the arm circumference
at a point midway between the olecranon and the
The prevalence of hypertension is increasing acromion, and the bladder length should cover
in childhood along with the increase in the 80–100 % of the circumference of the arm. Blood
­prevalence of obesity. Hypertension is ­diagnosed pressure should be determined in the resting state
if the mean systolic or diastolic blood pressure rather than in the excitement of entering the doc-
is above the 95th percentile for gender, age, and tor’s office, and measurements in the lying, sitting,
height on three or more occasions. The limit of and standing positions might be required if postural
stage 1 hypertension is a blood pressure between hypotension is suspected; pulse in each position
the 95th and 99th percentile plus 5 mmHg, should likewise be determined. A decrease in blood
while stage 2 hypertension is a blood pressure pressure of 10 mmHg or an increase in pulse rate of
over 5 mmHg above the 99th percentile plus 20 is considered significant as an indicator of pos-
5 mm of mercury. A new classification of pre-­ tural hypotension.
hypertension in childhood has been added and The family history must explicitly determine
defined as a mean systolic or diastolic blood whether close relatives have hypertension
pressure at or above the 90th percentile but (some will say their relatives do not have hyper-
below the 95th percentile. Pre-hypertension is tension while they are successfully treated
also diagnosed with blood pressures greater with ­medication, so the question should also be
than 120/80 mm of mercury. “do you take medication for hypertension”).
Evaluation of blood pressure may present Medication history must be obtained since stimulant
problems in some clinicians’ offices. The auto- medications may raise blood pressure as may
matic blood pressure device in frequent use may pseudoephedrine for allergies. Likewise, it is
not be as accurate as a manual sphygmomanom- important to determine if the individual smokes
eter, and the standards are derived from the man- or uses illicit drugs. Natural licorice can raise
ual sphygmomanometer method. If there is ever a blood pressure but not the candy version usually
question, the manual device must be used. Some found in the USA.
children may have white coat hypertension in Because elevated blood pressure may be sec-
which the blood pressure is elevated in the pres- ondary to another condition and may cause end-­
ence of the physician or in the clinicians’ office, organ complications, a phased approach to
while others may have masked hypertension in evaluation is required. Besides the standard eval-
which the blood pressure is normal in the office uations recommended for obesity including
but high in the ambulatory setting. In such cases, comprehensive metabolic panel and microalbu-
ambulatory 24-h blood pressure readings may be min determination in the urine, in addition,
invoked for definitive diagnosis. Ambulatory plasma renin activity and aldosterone determina-
blood pressure monitoring is recommended in tions, urinalysis and culture, CBC, and renal
patients with diabetes, chronic kidney disease, ultrasound are recommended. Echocardiogram
episodic hypertension, or autonomic dysfunc- to determine left ventricular mass and function is
tion. Coarctation of aorta is ruled out by mea- carried out if hypertension is discovered. In
surements of arm and leg blood pressures. selected cases suggesting pheochromocytoma or
The size of the cuff is of extreme importance, Cushing syndrome, evaluation of catechol-
and some children with obesity may require amines and glucocorticoid production is war-
remarkably large cuffs for accurate determinations. ranted (see Chap. 10).
348 13 Obesity

If the hypertension appears related to obesity, recommending initial use of angiotensin-­


lifestyle modification is always recommended as converting enzyme (ACE) inhibitor with calcium
is true in uncomplicated obesity although in the channel blockers next in line followed by diuretics
case of hypertension, a DASH (Dietary and beta-blockers; some recommend the use of
Approaches to Stop Hypertension) diet is also more than one agent in a refractory case. ACE
invoked. If the DASH diet is not successful, phar- inhibitors are teratogenic as are statins and con-
macologic treatment would be next with most traception or abstinence is required of

Table 13.2  Blood pressure levels for the 90th and 95th percentiles of blood pressure for boys aged 1 to 17 years by
percentiles of height
Age Systolic BP (mmHg) by percentiles height-> Diastolic BP (mmHg) by percentiles height->
(Year) BP % ile 5th 10th 25th 50th 75th 90th 95th 5th 10th 25th 50th 75th 90th 95th
1 90th 94 95 97 99 100 102 103 49 50 51 52 53 53 54
95th 98 99 101 103 104 106 106 54 54 55 56 57 58 58
2 90th 97 99 100 102 104 105 106 54 55 56 57 58 58 59
95th 101 102 104 106 108 109 110 59 59 60 61 62 63 63
3 90th 100 101 103 105 107 108 109 59 59 60 61 62 63 63
95th 104 105 107 109 110 112 113 63 63 64 65 66 67 67
4 90th 102 103 105 107 109 110 111 62 63 64 65 66 66 67
95th 106 107 109 111 112 114 115 66 67 68 69 70 71 71
5 90th 104 105 106 108 110 111 112 65 66 67 68 69 69 70
95th 108 109 110 112 114 115 116 69 70 71 72 73 74 74
6 90th 105 106 108 110 111 113 113 68 68 69 70 71 72 72
95th 109 110 112 114 115 117 117 72 72 73 74 75 76 76
7 90th 106 107 109 111 113 114 115 70 70 71 72 73 74 74
95th 110 111 113 115 117 118 119 74 74 75 76 77 78 78
8 90th 107 109 110 112 114 115 116 71 72 72 73 74 75 76
95th 111 112 114 116 118 119 120 75 76 77 78 79 79 80
9 90th 109 110 112 114 115 117 118 72 73 74 75 76 76 77
95th 113 114 116 118 119 121 121 76 77 78 79 80 81 81
10 90th 111 112 114 115 117 119 119 73 73 74 75 76 77 78
95th 115 116 117 119 121 122 123 77 78 79 80 81 81 82
11 90th 113 114 115 117 119 120 121 74 74 75 76 77 78 78
95th 117 118 119 121 123 124 125 78 78 79 80 81 82 82
12 90th 115 116 118 120 121 123 123 74 75 75 76 77 78 79
95th 119 120 122 123 125 127 127 78 79 80 81 82 82 83
13 90th 117 118 120 122 124 125 126 75 75 76 77 78 79 79
95th 121 122 124 126 128 129 130 79 79 80 81 82 83 83
14 90th 120 121 123 125 126 128 128 75 76 77 78 79 79 80
95th 124 125 127 128 130 132 132 80 80 81 82 83 84 84
15 90th 122 124 125 127 129 130 131 76 77 78 79 80 80 81
95th 126 127 129 131 133 134 135 81 81 82 83 84 85 85
16 90th 125 126 128 130 131 133 134 78 78 79 80 81 82 82
95th 129 130 132 134 135 137 137 82 83 83 84 85 86 87
17 90th 127 128 130 132 134 135 136 80 80 81 82 83 84 84
95th 131 132 134 136 138 139 140 84 85 86 87 87 88 89
From the The Fourth Report on the Diagnosis, Evaluation, and Treatment of High Blood Pressure in Children and
Adolescents U.S. DEPARTMENT OF HEALTH AND HUMAN SERVICES
National Institutes of Health
National Heart, Lung, and Blood Institute
NIH Publication No. 05-5267
Originally printed September 1996 (96-3790)
Revised May 2005
Comorbidities of Childhood and Adolescent Obesity 349

Table 13.3  Blood pressure levels for the 90th and 95th percentile of blood pressure for girls aged 1 to 17 years by
percentiles of height
Systolic BP (mmHg) by percentiles height-> Diastolic BP (mmHg) by percentiles height->
Age % ile 5th 10th 25th 50th 75th 90th 95th 5th 10th 25th 50th 75th 90th 95th
(Year) Ht
1 90th 97 97 98 100 101 102 103 52 53 53 54 55 55 56
95th 100 101 102 104 105 106 107 56 57 57 58 59 59 60
2 90th 98 99 100 101 103 104 105 57 58 58 59 60 61 61
95th 102 103 104 105 107 108 109 61 62 62 63 64 65 65
3 90th 100 100 102 103 104 106 106 61 62 62 63 64 64 65
95th 104 104 105 107 108 109 110 65 66 66 67 68 68 69
4 90th 101 102 103 104 106 107 108 64 64 65 66 67 67 68
95th 105 106 107 108 110 111 112 68 68 69 70 71 71 72
5 90th 103 103 105 106 107 109 109 66 67 67 68 69 69 70
95th 107 107 108 110 111 112 113 70 71 71 72 73 73 74
6 90th 104 105 106 108 109 110 111 68 68 69 70 70 71 72
95th 108 109 110 111 113 114 115 72 72 73 74 74 75 76
7 90th 106 107 108 109 111 112 113 69 70 70 71 72 72 73
95th 110 111 112 113 115 116 116 73 74 74 75 76 76 77
8 90th 108 109 110 111 113 114 114 71 71 71 72 73 74 74
95th 112 112 114 115 116 118 118 75 75 75 76 77 78 78
9 90th 110 110 112 113 114 116 116 72 72 72 73 74 75 75
95th 114 114 115 117 118 119 120 76 76 76 77 78 79 79
10 90th 112 112 114 115 116 118 118 73 73 73 74 75 76 76
95th 116 116 117 119 120 121 122 77 77 77 78 79 80 80
11 90th 114 114 116 117 118 119 120 74 74 74 75 76 77 77
95th 118 118 119 121 122 123 124 78 78 78 79 80 81 81
12 90th 116 116 117 119 120 121 122 75 75 75 76 77 78 78
95th 119 120 121 123 124 125 126 79 79 79 80 81 82 82
13 90th 117 118 119 121 122 123 124 76 76 76 77 78 79 79
95th 121 122 123 124 126 127 128 80 80 80 81 82 83 83
14 90th 119 120 121 122 124 125 125 77 77 77 78 79 80 80
95th 123 123 125 126 127 129 129 81 81 81 82 83 84 84
15 90th 120 121 122 123 125 126 127 78 78 78 79 80 81 81
95th 124 125 126 127 129 130 131 82 82 82 83 84 85 85
16 90th 121 122 123 124 126 127 128 78 78 79 80 81 81 82
95th 125 126 127 128 130 131 132 82 82 83 84 85 85 86
17 90th 122 122 123 125 126 127 128 78 79 79 80 81 81 82
95th 125 126 127 129 130 131 132 82 83 83 84 85 85 86
From the The Fourth Report on the Diagnosis, Evaluation, and Treatment of High Blood Pressure in Children and
Adolescents U.S. DEPARTMENT OF HEALTH AND HUMAN SERVICES
National Institutes of Health
National Heart, Lung, and Blood Institute
NIH Publication No. 05-5267
Originally printed September 1996 (96-3790)
Revised May 2005

reproductive-­aged girls. Long-term studies are Liver Disease


lacking, but there is some evidence that damage
will not progress or in some cases reverse with Nonalcoholic fatty liver disease is now the most
appropriate therapy. common cause of chronic hepatic disease in child-
Blood pressure standards are found in hood and adolescence and may be seen as young
Tables 13.2 and 13.3. as 3 years of age. It starts with fatty infiltration of
350 13 Obesity

the liver (steatosis) and progresses to steatohepati- microbiome and polyunsaturated fatty acids
tis and can further progress to fibrosis and cirrho- although it is premature to recommend these for
sis; cirrhosis may lead to the development of liver therapy at present. There is some information to
cancer. NAFLD was found in about 10 % of chil- demonstrate beneficial effects of bariatric surgical
dren in autopsy study of all deaths in San Diego treatment of extreme obesity on NAFLD.
and in about 40 % of children with obesity in that
study. Children with severe obesity may have a
prevalence of NAFLD as high as 80 %. NAFLD is Obstructive Sleep Apnea
now considered as the hepatic manifestation of
the metabolic syndrome. Remarkably although The Institute of Medicine calls sleep disorders
obesity rates are relatively similar in African- “an underrecognized public health problem.”
American and Hispanic-­American children, the Obesity affects sleep, and abnormal sleep affects
NAFLD rate is highest in Hispanic children and metabolism.
lowest in African-­American children. OSA is characterized by chronic partial upper
The diagnosis of NAFLD is problematic as airway obstruction with intermittent complete
liver enzyme (ALT) determination has poor speci- airway obstruction that affects sleep and ulti-
ficity and sensitivity; elevated values are > 25 IU/L mately has major effects on health. Most but not
in boys and > 22 IU/L in girls (may be lab depen- all individuals with OSA snore. Diagnostic tech-
dent). Ultrasound studies can show fat in the liver niques include parent reports, parental question-
but not fibrosis, leaving liver biopsy as the only naires on sleep habits, audio or video recording
“gold standard,” but few children will have this of child sleeping, and, considered the gold stan-
procedure, so proxy measures are used. During dard, polysomnography. With polysomnography,
this epidemic of obesity, any child who has liver the number of apnea hypoxemia events per hour
disease from an etiology other than NAFLD can expressed as the AHI is used for diagnosis. An
also be obese. Thus, it is important for the practi- AHI higher than 5 is diagnostic of mild sleep
tioner to consider and eliminate other liver dis- apnea, greater than 15 for moderate sleep apnea,
eases such as hepatitis B and C, autoimmune and greater than 30 for severe sleep apnea.
hepatitis, drug-induced liver injury, Wilson’s dis- Obesity is a risk factor for OSA with increas-
ease, alpha 1-antitrypsin deficiency, inborn errors ing risk with increasing BMI. OSA leads to CO2
of fatty acid or carnitine metabolism, peroxisomal retention, hypoxia, and right ventricular hyper-
disorders, lysosomal storage disorders, and cystic trophy and failure and pulmonary hypertension.
fibrosis before diagnosing NAFLD. This also is potentially associated with prolonga-
There appears to be a multifactorial etiology tion of the QTc interval and ventricular arrhyth-
of NAFLD. Elevated triglycerides reach the liver mias. The obesity hypoventilation syndrome or
and deposit ectopic fat; inflammatory cytokines Pickwickian syndrome, characterized by
affect the liver and rise in obesity, and even the hypoventilation, somnolence, CO2 retention,
gut microbiome plays a role. A gene related to hypoxia, polycythemia, right ventricular hyper-
NAFLD is patatin-like phospholipase domain-­ trophy and failure, pulmonary embolism, and
containing 3 (PNPLA3) gene variant I148M. even sudden death, is an extreme example of this
Treatment of NAFLD starts, as in the treatment process noted more frequently in older literature.
of obesity, with lifestyle modification. Although Pulmonary hypertension may result from OSA.
some medications have shown promise but ulti- Disordered sleep is a risk factor for glucose
mately failed to be more beneficial than lifestyle intolerance and weight gain in children and
modification (these include vitamin E, metformin, adults. Limited sleep increases appetite and
and ursodeoxycholic acid), there are still some caloric intake. Sleep quality should be investi-
promising approaches. Beneficial effects have been gated by history and sleep hygiene advice pro-
found with the use of probiotics to change the gut vided if necessary.
Comorbidities of Childhood and Adolescent Obesity 351

Dermal Findings in MC4R mutation. A short child with obesity has


a greater likelihood of having a definable endo-
Acanthosis nigricans manifest as dark pigmenta- crine disorder such as hypothyroidism, Cushing
tion on the back of the neck or other flexural syndrome, hypopituitarism or growth hormone
surfaces, with thickening and even papilloma
­ deficiency, and pseudohypoparathyroidism, and
­formation in more severe cases. This is a physical these will generally have a delayed bone age.
indication of insulin resistance. Intertrigo and
furunculosis may independently develop in skin
folds and increase pigmentation as well. Earlier Puberty
There is a correlation between obesity and
type 2 diabetes mellitus and the development of There is a trend to earlier puberty recently, and in
psoriasis. girls, this is often linked to increasing BMI.
Menarche and the other aspects of pubertal devel-
opment are found earlier than their peers. Early
Orthopedic Conditions menarche raises the risk of breast cancer later in life
so that earlier puberty is a cause for concern. This
Obesity in childhood and adolescence leads to a problem is discussed in more detail in Chap. 9.
host of orthopedic conditions mostly related to
weight-bearing extremities.
Slipped capital femoral epiphysis (SCFE) Enuresis
occurs when the proximal femoral metaphysis sep-
arates from the epiphysis of the femoral head. This Enuresis is found more frequently in children
condition most often occurs in obese individuals with obesity. It can occur without sleep apnea
and also is found in hypothyroidism or in the rapid although studies vary in this relationship.
growth of growth hormone treatment. Usually hip,
leg, or foot pain develop and the child is noted to be
waddling during ambulation. The incidence of Cholelithiasis
slipped capital femoral epiphysis is increasing
along with the incidence of obesity in childhood. Cholesterol cholelithiasis is associated with pedi-
Surgery is required for treatment of SCFE. atric obesity, and while rare, the prevalence is ris-
Blount disease develops from abnormal growth ing. Cholelithiasis is also found with rapid weight
of the medial part of the proximal tibial physis and loss, and prophylactic treatment with oral
results from increased weight on the proximal ­ursodeoxycholic acid may be considered after
tibia. This leads to abnormal gait, discrepancy in bariatric surgery. There is the added risk of the
limb length, and a tendency toward the develop- development of pancreatitis.
ment of arthritis. There are two forms, infantile
and adolescent Blount disease. Characteristically,
surgery is the mode for treatment, but the infantile Asthma
form might be amenable to nonsurgical treatment.
Low back pain is also more common in ado- Asthmatic children may have limited physical
lescents with obesity. activity that fosters the development of obesity.
On the other hand, obesity in children causes pre-
existing asthma to worsen.
I ncreased Growth and Advanced
Skeletal Development
Pseudotumor Cerebri
Advancement of bone age and tall stature are
common in most otherwise normal but children Pseudotumor cerebri is more common in obesity
with obesity; bone age advancement is described and presents with headaches. Clinical diagnosis
352 13 Obesity

requires ophthalmoscopy to detect papilledema, For symptoms suggestive of obstructive sleep


an otherwise normal neurologic exam, and MRI apnea, a sleep study will be performed. For suspi-
to detect evidence of normal central nervous sys- cion of hypertension, ambulatory BP monitoring
tem parenchyma, absence of hydrocephalus or is invoked. For orthopedic symptoms, extremity
masses, normal spinal fluid composition, and exam and radiographs are obtained.
elevated cerebrospinal fluid pressure. Some Thyroid-function tests (free T4 and TSH) are
patients will have pseudotumor cerebri syn- of low yield as hypothyroidism should not cause
drome without papilledema which presents a a significant level of obesity, but parents often
diagnostic challenge although the other criteria insist on thyroid testing, having obtained infor-
noted above must be satisfied in this situation as mation from the Internet suggesting that it is a
well. Weight loss will likely be curative, but cause of obesity.
shunting may be necessary in some cases. Genetic testing for one of the single gene muta-
tions listed above is reserved for those children
with the onset of obesity before 5 years of age,
Psoriasis have hyperphagia, demonstrate physical findings
of genetic syndromes or who have extreme obesity
Psoriasis is an inflammatory disease which has in their family. Developmental delay is found in
been linked to obesity in adults and in children, some of the genetic obesity conditions but not all.
but no cause-and-effect relationship is yet
established.
Treatment

It is indeed hard to believe, but some families and


 aboratory Evaluation of Childhood
L patients do not connect the intake of french fries
Obesity and high-calorie drinks with the development of
obesity. Education rarely is sufficient to change
The degree of investigation is dependent on behavior, but it has to be the starting point of an
the BMI and the presence of risk factors. encounter. Of course a history of increased caloric
Recommendations are based upon expert opinion intake and/or decreased activity will be found in
tempered by the prevalence of comorbidities of many subjects with obesity; some may already be
obesity. quite active in sports or other activities and may
If the BMI for age is in the 85th to 94th appear to follow a reasonable diet. In these cases,
­percentile with no risk factors, a fasting lipid pro- often portion sizes or second helpings are respon-
file is obtained. sible for the excess caloric intake. These children
If risk factors are present, and this is the case may still require changes in behavior to decrease
in most situations, a comprehensive chemistry caloric intake and increase in energy expenditure
panel with fasting glucose and an alanine amino- in a program of lifestyle modification. There is
transferase (ALT) measurement is obtained. only one approved drug for the treatment of child-
HgbA1c is measured. Although obesity is an insu- hood and adolescent obesity, and surgical approach
lin resistance syndrome, a fasting insulin level to obesity is quite a significant step, so lifestyle
does not indicate the likelihood of type II diabe- modification must remain the basis of treatment.
tes or any other complications and is generally The goal of lifestyle modification is not to
not recommended. Insulin levels are used for make a stick-thin child but a healthier child.
clinical studies but not in individual patients. There is metabolic benefit from small changes in
If BMI is equal to or greater than the 95th per- BMI whatever treatment program brings about.
centile, all of the listed tests are recommended Thus a decrease in 0.5 BMI units or in 8 %
irrespective of complications. decrease in BMI, depending upon the study, has
If appropriate historical features suggest par- been shown to improve metabolic balance and
ticular problems, additional tests are required. insulin resistance in children.
Laboratory Evaluation of Childhood Obesity 353

It must be remembered that many families portion sizes and processed foods often with salt
have many psychosocial issues in their life over and high-fructose corn syrup added. An impor-
and above the obesity of the child. The families tant goal is not to make the child feel that he or
may come from lower socioeconomic status or she is always hungry which will sabotage any
live in dangerous neighborhoods. All of these attempts at moderate diet. Unfortunately as an
issues impair their ability to follow the sugges- obese individual loses weight, it becomes harder
tions we may offer in a clinic visit. Understanding to lose more weight, and a plateau is reached. The
this situation may improve communication with reduction of calories for weight maintenance or
the families. weight loss in the obese person is greater than the
reduction of calories for weight maintenance or
weight loss in a nonobese patient, making caloric
Lifestyle Modification deficits all the harder to achieve.
The primary dietary treatment for any child
In clinical encounters, no provider should use the requiring weight control is decreased caloric
terms obesity or overweight but rather should intake (about 500-calorie/day decrease in older
discuss with the family healthy habits including children) but not so much as to stunt statural
dietary intake and activity levels. The aim in life- growth. The provider will encounter numerous
style modification is to decrease caloric intake different cultures which have their own forms of
and to increase activity. There are sometimes high caloric intake. Every family must be treated
obvious changes which can be discussed with the with respect for their cultural backgrounds, but
family. For instance, one goal can be the elimina- usually some traditional food can be invoked to
tion of sugar-sweetened beverages such as colas exclude others. For the Hispanic families, one
by replacing it with water. Intake of juice should can discuss decreasing cooking food with lard or
adhere to the recommendations of the Academy frying food and replacing these methods with
of Pediatrics which suggest that 1- to 6-year-old cooking with vegetable oil and steaming. Some
children should have only 120–150 mL of juice traditional food such as cactus carry low caloric
per day and 7–18-year-olds limited to 240– content, while others such as rice and beans may
360 mL per day. Water is the best oral intake for contain more. An Asian diet often can include
a child with obesity, but if flavor is craved, flavors many vegetables cooked in vegetable oil to the
can be added with products such as Crystal Light exclusion of fried foods or large servings of meat.
which contains no calories and has various fruit These adjustments must be suggested in a
flavors. Substituting diet drinks for high-calorie respectful manner. Suggestions in English and
high-­fructose corn syrup-laden sugar-sweetened Spanish and handouts for Anglo and Hispanic
beverages is not an answer as there is evidence families are found at http://win.niddk.nih.gov/
that drinking diet drinks not only does not help to index.htm and additional resources including
stop weight gain but actually can also lead to some for Asian families at HDL http://www.eat-
weight gain. A source of calcium must remain r i g h t . o rg / H e a l t h P r o f e s s i o n a l s / c o n t e n t .
even if the percent of fat in milk is decreased. aspx?id=250. Changes are encouraged by using
Schools should be encourage to offer water rather motivational interviewing discussed below in this
than juice or sugar sweetened beverages in the chapter. Change takes time, and it is essential to
cafeteria or vending machines. negotiate a reachable goal during clinical encoun-
Approaches used in the adult can be beneficial ters. Eliminating or cutting down on sugar-sweet-
in children. Emphasizing eating breakfast rather ened beverages might be a goal for one visit and
than skipping it is an important goal. When adults eating one more serving of vegetable per day for
actually write down what they are eating, it another. Figure 13.3 provides a starting point for
focuses their attention on the content of the food, increasing the use of the MyPlate model in
and teenagers may benefit from this as well. English and Spanish as other reasonable goals.
Decreased episodes of eating at restaurants or The family must be involved in the approach to
fast-food outlets will decrease exposure to large younger children.
354 13 Obesity

Increased energy expenditure aims to decrease which the family at least plans to make changes in
sedentary activity rather than forcing vigorous unhealthy behaviors; this is followed by action in
exercise, which will rarely be maintained. which the changes are actually made in unhealthy
Increased activity benefits the least mobile behaviors; the last stage is maintenance of the
­children, the most in terms of their metabolism. changes that have been made. Characteristically
Even five episodes of 20 min of exercise per an individual and family will move up and down
week can improve insulin sensitivity and decrease these five steps, so the provider might be able to
liver enzyme abnormalities over a period of support progress toward maintenance more effec-
months. This may occur even without significant tively by continued motivational interviewing.
weight loss since muscle weighs more than fat A practitioner might reinforce positive behav-
tissue, and as muscle tissues build up, weight ior by lauding the family for any positive activity
may remain stable. The use of video games that such as exercise or eating vegetables. The pro-
encourage physical activity may be invoked, and vider might then ask what is more negative such
family walks can be promoted. While many as excessive caloric drinks or sedentary time
schools are far from the student’s house, walking which might be received more positively since it
to school with family members or using a “walk- follows a positive statement. The provider might
ing school bus” in which neighbors band together then ask what negative behavior the family might
to walk children to the school increases activity want to talk about. Taking the lead from the fam-
levels and is a very beneficial manner. Gym ily in this way allows better communication. The
classes are not required in many schools at the provider might talk about the positives and nega-
present time, so extracurricular physical activity tives of the unhealthy behavior such as high-­
such as sports teams is important. Determination calorie sugar-sweetened beverages. After asking
of the child’s living environment is of great for permission, the provider might then provide
importance. Is a child left at home as a “latchkey information about the unhealthy behavior in
child” having free access to chips and television order to enhance the message. The provider
while parents work? Is it possible to have the might then ask on a 1–10 score how important
child go to an afterschool activity or Boys and this change in behavior is to the family and how
Girls Club in the neighborhood? Is it possible to confident they are in making the changes. If they
have some kind of exercise device in the house or choose a low number, the provider might ask
apartment if the outside environment is unsafe? why the number is low in trying to guide the
Standard approaches to the recommendation family into a more positive outlook. Alternately,
of lifestyle changes to families are often ineffec- the provider might ask why the number is high
tive unfortunately. Motivational interviewing is such as 9 or 10, and the provider might comple-
suggested as a more effective manner of motivat- ment the family on how important this decided
ing families to make changes. In most common change is to them. The meeting then ends with a
medical communications, the provider directs summary to go over everything that was dis-
patient and family activities while motivational cussed in order to reinforce the messages
interviewing aims to guide families into changes developed.
using appropriate communication techniques and A group program of education and family sup-
their outlooks. The stages of change model is not port is superior to individual sessions for children.
accepted by all authorities but may nonetheless Family therapy is used with success in some cen-
be useful in clinical interactions when evaluating ters. Group programs may follow the Traffic
and motivating families. In this concept, there are Light, Shapedown, or our UCDMC Fit-­Kid mod-
five stages: One is pre-contemplation when a els. In the youngest children, the parents solely
child’s weight is not even raised in the families’ determine the diet for the child and must play an
consciousness; the second is contemplation which important role in the counseling, whereas the
they are at least thinking about the fact that the message might be more focused on the subjects
child may be ­overweight or that his or her habits during the adolescent years. Community involve-
might be unhealthy; the next step is preparation in ment in the approach to the obesity epidemic is
Laboratory Evaluation of Childhood Obesity 355

important in a global approach to the problem. day intake, including 1.5–2.5 g high-quality pro-
Unfortunately, only a few programs show long- tein per kilogram ideal body weight per day (sup-
term success, as constant reinforcement of healthy plied from lean meats, poultry, and fish) but only
habits is necessary and not often available; obe- 20–40 g/day of carbohydrate; daily vitamin and
sity should be considered as a chronic disease that mineral supplementation and encouragement to
requires continuous management. consume more than 1,500 mL free water are addi-
An essential point for anyone caring for chil- tional important features. A PSMF is carried out
dren with obesity is a sense of empathy and a usually for no more than 12 weeks, with daily
supportive manner that does not further decrease monitoring for urinary ketones to demonstrate
the child’s self-esteem; if providers cannot mus- adherence to the low-­caloric plan. The risks of
ter such feelings, they should not be involved PSMF ­regimens include cholelithiasis; hyperuri-
with affected children or families. cemia; decreases in serum proteins, including
The goals of weight management are based on transferrin, retinol-binding protein, and comple-
age, BMI, and complications (Fig. 13.2). If the ment 1C; orthostatic hypotension; halitosis; and
child can grow into the weight, stopping further diarrhea. Although long-­term weight control may
weight gain is the goal, but if any weight-based not result, lessening of the initial medical compli-
complications are noted, weight loss is needed. cations may be achieved. This technique has been
Thus in the child younger than 2 years, if the BMI suggested as an initial step before bariatric surgery
is less than 95 %, dietary counseling is recom- is performed. It must be performed by an experi-
mended, but if the BMI is greater than 95 %, spe- enced team.
cialty consultation is suggested. In a child or early
adolescent with a BMI of 85–94 % and no com-
plications, or in a child younger than 7 years who Medication
has a BMI greater than 95 % but no complica-
tions, weight maintenance is recommended, as Orlistat is the only medication approved for obe-
the child will grow into the weight if no more is sity in childhood. Orlistat is an intestinal lipase
gained. However, if the BMI is greater than 95 % inhibitor that decreases fat intake and increases
at older than 7 years or if complications are found, the elimination of dietary fat. Side effects include
weight loss rather than weight maintenance is diarrhea and flatus if significant fat remains in the
required. Specialty consultation becomes prob- diet, so this becomes a form of aversion therapy
lematic in many locations as this type of a pro- if diet is not first modified. The unpleasant side
gram is usually available at academic medical effects may make teenagers cease to take this
centers or large hospital systems rather than small medication.
towns. With the growth of telemedicine, consulta- Metformin, a biguanide that decreases hepatic
tion can be offered from a distance to many rural release of glucose and decreases insulin resis-
communities. Alternatively, expert education for tance, is approved for diabetes in children as
local practitioners through telemedicine by dis- young as 10 years of age but not for weight
tant consultants might increase the abilities of the ­control. Nonetheless, several studies have shown
community to deal with the obesity problems. a modest decrease in BMI (about 1.5 kg/m2) with
A protein-sparing modified fast (PSMF) has the use of this medication so that in selective
been used in children with severe, often life-­ cases of extreme obesity, it may have benefited
threatening, consequences of obesity (e.g., sleep even though it is used off label. While metformin
apnea) in which rapid weight loss is indicated, if can cause abdominal distress, usually temporary,
no other method appears applicable, but must be it is generally considered safe. Rare individuals
administered only with an experienced practitio- will have elevations of creatinine kinase and
ner because of inherent dangers. There is very lit- muscle pain or rhabdomyolysis. If so, metformin
tle recent literature on this method. The PSMF is a must be immediately stopped. In adults, there is a
medically supervised (an experienced clinician tendency toward lactic acidosis. Vitamin B12
must direct the therapy) program of 600–800 kcal/ deficiency is a risk with metformin treatment so
356 13 Obesity

daily vitamin intake is advised with metformin suggestive of the adult literature in which weight
treatment. Exenatide is an injectable glucagon- loss continues for years after the procedure.
like peptide-­1 (GLP-1) agonist approved for the Adjustable gastric banding is approved for patients
treatment of type II diabetes in adults. It pro- over 18 years of age but not for those that are
motes a feeling of fullness and decreases appe- younger. Adjustable gastric banding decreases the
tite, leading to a degree of weight loss. A few diameter of the stomach at the caudal region,
clinical studies in children suggested that a restricting the amount of food ingested. An inflat-
decrease in BMI of 4 % is possible. Exenatide is able band is placed around the antrum of the stom-
not approved by the FDA for use below 16 years ach; the band can be inflated through a port
of age. Extended-­release exenatide is associated subcutaneously. There is more limited information
with medullary thyroid carcinoma in rodents. on this procedure in teenagers and due to compli-
cations is rarely used anymore. A more recently
developed procedure, vertical sleeve gastrectomy
Surgery which involves stapling or surgically removing
most of the stomach, leaving a tube connecting the
The results of surgical treatment for obesity in esophagus with the duodenum, holds promise, but
adolescents are limited compared to data from there is less data in adolescents with this proce-
adults, but short- and intermediate-term outcomes dure. Further data is required on the effects of sur-
are accumulating. Consensus guidelines suggest gery in teenagers, and ongoing national studies
that an adolescent with a BMI greater than 40 with will direct the clinician in the future.
the presence of a severe comorbidity or those with There are guidelines for the choice of appropri-
a BMI greater than 50 with a less severe comorbid- ate patients to undergo surgery. If a girl is 13 years
ity may be candidate for bariatric surgery assum- old or a boy is 15 years of age, bariatric surgery
ing that an appropriate program is available. can be considered according to recent guidelines if
Anyone considering bariatric surgical treatment the adolescent has failed 6 months of organized
for adolescent obesity should ensure that the child attempts at weight management (determined by
is enrolled in a long-term outcomes study so that their primary care provider), has attained or nearly
the much needed data on the best approach to attained physiologic maturity, is severely obese
complications and the advantages of surgical treat- (BMI > 40) with serious obesity-related comor-
ment can be determined. Surgery requires a team bidities or has a BMI of >50 with less severe
to screen appropriate patients and support the pro- comorbidities, demonstrates commitment to com-
cess long into the postoperative period and is best prehensive medical and psychologic evaluations
carried out in the environment of a clinical study both before and after surgery, agrees to avoid preg-
rather than in a local hospital and only at an ado- nancy for at least 1 year postoperatively and will
lescent bariatric surgery center of excellence. not or is not breast feeding, is capable of and will-
The longest-term experience in surgical ing to adhere to nutritional guidelines postopera-
approaches to adolescent obesity involves the tively, provides informed assent to surgical
Roux-en-Y gastric bypass (RYGB) whereby a treatment, demonstrates decisional capacity, and
small pouch is created, separating it from the rest has a supportive family environment. Severe com-
of the stomach and allowing food to exit the stom- plications are type 2 diabetes mellitus, OSA, and
ach directly into the middle section of the small pseudotumor cerebri. Less severe complications
intestine. This leads to restriction in the amount of are hypertension, dyslipidemias, nonalcoholic ste-
food that will be eaten and changes the endocrine atohepatitis, venous stasis disease, significant
milieu by decreasing ghrelin secretion from the impairment in activities of daily living,
stomach since less of the stomach is exposed to ­intertriginous soft tissue infections, stress urinary
ingested food. Patients may lose 10 BMI points or incontinence, gastroesophageal reflux disease,
approximately 35 % of their weight in the first 6 weight-related arthropathies that impair physical
months. There is limited long-term data, but it is activity, and obesity-related psychosocial distress.
Laboratory Evaluation of Childhood Obesity 357

Prevention healthy activity level, and everyone including the


parents and the siblings can benefit from this life-
With one-third of the childhood and teenage pop- style modification.
ulations overweight or obese, it is clear that pre-
vention must take a preeminent role in addressing
this epidemic. The encouraging news is that the  xpose Children to Healthy Food
E
prevalence of obesity in children and adolescents in a Young Age
is not increasing in most US groups demonstrat-
ing the likelihood that some of the public health Children are born with a taste for sweet and
measures that have been instituted are having an salt and an aversion to bitter and sour and
effect. Prevention includes anticipatory guidance ­demonstrate a tendency to avoid new foods (neo-
that providers may give to families and patients. phobia); the desire for fat or oil, spice (other than
The important issues of city planning and archi- salt), and fat may be learned at an early age. Thus
tectural design may not seem to be within the a child will initially spit out green vegetables; if
purview of a pediatric practitioner, but these the parent gives up such trials of healthful food,
areas require advocacy to develop safe play areas resorting to sugary or salty, initially more palat-
and structures and accessible buildings that have able substitutes, the child may lose the chance to
stairs easily available rather than the omnipresent acquire long-term healthful dietary habits. But
elevators. Other countries limit advertising to just as a lenient approach to diet may be harmful,
children. This is an important area as children are overly restricting food choices to the point at
exposed to foods totaling as much as 20,000 calo- which the child cannot learn to regulate their
ries per hour while watching television on intake, conversely, also may foster the develop-
Saturday a.m. The USA does not limit advertis- ment of obesity.
ing to children, and this is another appropriate
target for advocacy.
Encourage Breast-Feeding

Encourage Parental Modeling For many reasons, breast-­ feeding should be


encouraged in all children. However, breast-
The major role model of small children is of feeding has not been conclusively demonstrated
course parents. Parents having excessive por- to decrease the risk for obesity. Modern research
tions, taking multiple servings, or “grazing” into the gut microbiome demonstrates that the
throughout the day interspersing such activity gastrointestinal flora of children delivered by
with sugar-sweetened beverages and a sedentary C-section differs from those delivered vaginally,
lifestyle may have an effect in children as young differs from children raised on formula rather
as toddler years in developing bad habits. Some than breast milk, and as stated above, differs
parents have adverse habits that they refuse to from those who are obese with those who are
change while professing to want the children to lean. Breast-feeding therefore is an important
develop healthier habits. The parent must model aspect of developing a normal microbiome
appropriate behaviors in diet and activity, and which is more likely to lead to a healthier weight.
clearly the family must be the model for the child Further research progresses in evaluating probi-
and must participate in the treatment along with otic treatment of infants who were raised on for-
the child. Sometimes only one child in the family mula to restore the gut flora to the normal
is overweight or obese, while the others are thin. condition of a breast-fed baby.
Parents may say that they cannot treat the whole Bottle-fed babies may have increased weight
family the same way just because of one child. as they may grow up less likely to regulate the
Indeed they can because the needs of the child intake by their own choice. Formula or breast
with obesity are a healthy dietary intake and milk may belong in a bottle, but juice and soft
358 13 Obesity

drinks do not: It is remarkable to note that there in a sedentary manner, but the best situation
are baby bottles for sale that have soft drink would be to have the child in a guided after-
insignias on them, and some parents believe that school program which encourages good nutri-
they should be filled with soft drinks even during tion, increased activity, and studying.
the first year. Bottles should be used only up to
about 1 year, if used at all. Bedtime bottle-­feeding  ecrease “Screen” Watching
D
is particularly discouraged, as this often leads to Television viewing is the most proven modifi-
formation of dental caries as well as increased able factor linked to the development of obe-
caloric intake. sity. The average US child watches more than
28 h of television or watching other electronic
screens per week. Television viewing decreases
Decrease or Eliminate the Intake beneficial physical activity and socialization.
of Sugar Sweetened Beverages Exposure to television advertising that features
and Limit the Intake of Juice highly caloric foods may affect the diet. Some
success has occurred with a curriculum aimed
An increase in each soda consumed per day in to decrease television viewing. The Academy
early teenage years leads to doubling the likeli- of Pediatrics recommends limiting screen time
hood of developing obesity. Indeed in a rough to no more than 2 h per day. Since homework
sense, a daily serving of 12 ounces of sugar-­ now requires screen use a goal is to balance
sweetened carbonated beverage can lead to a 20 lb necessary screen exposure with activity.
weight gain after 1 year, all things being equal
(this is only illustrative as the actual weight equiv-
alence will vary). Juice may seem more healthful Modify the Diet
than sugared drinks, but 8 oz of juice contains 100
calories, just as 8 oz of soda! The best exposure to A small indiscretion in caloric intake adds up.
fruit is in the actual fruit rather than juice from the Consistent changes in diet toward decreased cal-
fruit. It is difficult to consume enough fruit to ories and increased fruit and vegetables will lead
cause weight gain, but when the fruit is rendered to a beneficial effect. A child cannot exercise off
into juice, it is very easy to drink excessive calo- a highly caloric children’s fast-food meal (con-
ries. Parents should limit juice and encourage tains 750–1,400 calories or more) in the gym or
water intake. Milk is an important source of vita- on the exercise bicycle. Approach to diet is noted
min D and calcium, but low-fat or skim milk is above, but there is no substitute for a dietician
best in the approach to overweight or obesity. being involved in counseling. Refer to Figs. 13.3
and 13.4 for examples of the use of the USDA
MyPlate model.
Increase Public Safety

If a child is not allowed to leave the house for Decrease Sedentary Time
fear of injury, the child will eliminate vigorous
activity and may stay in front of the television Decreasing the time spent sitting or being driven
set all day with servings of chips or sugar-sweet- around by replacement of this time with activities
ened beverages easily available. “Latchkey” kids such as walking or playing is beneficial. No child
are an unfortunate phenomenon in this economic will suddenly change from sedentary habits to
environment where both parents have to work, vigorous activity, so aim for sustainable small
sometimes at multiple jobs, when childcare or changes. Many children simply watch others par-
after-school programs are not available. It may ticipate in gym class, so substitution with fun
be possible for a child restricted to the house to activities rather than competitive activities may
use a video game that requires activity rather increase participation by all and lead to habits
than simply watching television or while sitting that might be retained. Since schools are at a
Laboratory Evaluation of Childhood Obesity 359

Fig. 13.4 (a and b) An example of goal setting in English and in Spanish for the introduction of the MyPlate model
into daily life. This tool may be used in motivational interviewing. Courtesy of Marilyn Townsend UC Davis with
permission
360 13 Obesity

d­ istance from the location where children live 6. Nogueira RC, Weeks BK, Beck BR. Exercise to
improve pediatric bone and fat: a systematic review
and children may be too young and the environ-
and meta-analysis. Med Sci Sports Exerc. 2014;
ment may be too dangerous for them to walk 46(3):610–21.
alone to school, parents have organized “walking 7. Ng M, Fleming T, Robinson M, Thomson B, Graetz
school buses” where parents walk along with the N, Margono C, Mullany EC, Biryukov S, Abbafati C,
Abera SF, Abraham JP, Abu-Rmeileh NME, Achoki T,
children, keeping them safe while monitoring the
AlBuhairan FS, Alemu ZA, Alfonso R, Ali MK, Ali R,
progress in bringing them to school. Thus the Guzman NA, Ammar W, Anwari P, Banerjee A,
children have walked moderate distance and Barquera S, Basu S, Bennett DA, Bhutta Z, Blore J,
increased energy expenditure and saved the envi- Cabral N, Nonato IC, Chang J-C, Chowdhury R,
Courville KJ, Criqui MH, Cundiff DK, Dabhadkar
ronment from car or bus emissions.
KC, Dandona L, Davis A, Dayama A, Dharmaratne
Conclusion: The best we can say about the obe- SD, Ding EL, Durrani AM, Esteghamati A, Farzadfar
sity epidemic is that it seems to have leveled off. F, Fay DFJ, Feigin VL, Flaxman A, Forouzanfar MH,
We cannot be proud that one-third of the adults in Goto A, Green MA, Gupta R, Hafezi-Nejad N,
Hankey GJ, Harewood HC, Havmoeller R, Hay S,
the USA are obese and that 33 % of ­children are
Hernandez L, Husseini A, Idrisov BT, Ikeda N, Islami
obese or overweight. If the factors that changed in F, Jahangir E, Jassal SK, Jee SH, Jeffreys M, Jonas
the environment in which we raise children con- JB, Kabagambe EK, Khalifa SEAH, Kengne AP,
tributed to the epidemic of obesity, changing such Khader YS, Khang Y-H, Kim D, Kimokoti RW, Kinge
JM, Kokubo Y, Kosen S, Kwan G, Lai T, Leinsalu M,
environmental factors should decrease the preva-
Li Y, Liang X, Liu S, Logroscino G, Lotufo PA, Lu Y,
lence of obesity. The steps outlined in this chapter Ma J, Mainoo NK, Mensah GA, Merriman TR,
are in some cases supported by compelling evi- Mokdad AH, Moschandreas J, Naghavi M, Naheed A,
dence, while others are suggestive of the common Nand D, Narayan KMV, Nelson EL, Neuhouser ML,
Nisar MI, Ohkubo T, Oti SO, Pedroza A, Prabhakaran
sense approach to the problem. This is going to be
D, Roy N, Sampson U, Seo H, Sepanlou SG, Shibuya
a long-term project for our country and the world. K, Shiri R, Shiue I, Singh GM, Singh JA, Skirbekk V,
It is likely to take decades and maybe generations Stapelberg NJC, Sturua L, Sykes BL, Tobias M, Tran
due to the epigenetic phenomena that affects sub- BX, Trasande L, Toyoshima H, van de Vijver S,
Vasankari TJ, Veerman JL, Velasquez-Melendez G,
sequent generations.
Vlassov VV, Vollset SE, Vos T, Wang C, Wang X,
Weiderpass E, Werdecker A, Wright JL, Yang YC,
Yatsuya H, Yoon J, Yoon S-J, Zhao Y, Zhou M, Zhu S,
Lopez AD, Murray CJL, Gakidou E. Global, regional,
Suggested Readings and national prevalence of overweight and obesity in
children and adults during 1980–2013: a systematic
analysis for the Global Burden of Disease Study 2013.
1. Yanovski SZ, Yanovski JA. Long-term drug treatment
Lancet. 2014;384(9945):766–81.
for obesity: a systematic and clinical review. JAMA.
8. Kelly AS, Dengel DR, Hodges J, Zhang L, Moran A,
2014;311(1):74–86.
Chow L, Sinaiko AR, Steinberger J. The relative con-
2. Skinner AC, Skelton JA. Prevalence and trends in
tributions of the abdominal visceral and subcutaneous
obesity and severe obesity among children in the
fat depots to cardiometabolic risk in youth. Clin Obes.
United States, 1999–2012. JAMA Pediatr.
2014;4(2):101–7.
2014;168(6):561–6.
9. Katzmarzyk PT, Barlow S, Bouchard C, Catalano PM,
3. Reyman M, Verrijn Stuart AA, van Summeren M,
Hsia DS, Inge TH, Lovelady C, Raynor H, Redman
Rakhshandehroo M, Nuboer R, de Boer FK, van den
LM, Staiano AE, Spruijt-Metz D, Symonds ME,
Ham HJ, Kalkhoven E, Prakken B, Schipper
Vickers M, Wilfley D, Yanovski JA. An evolving sci-
HS. Vitamin D deficiency in childhood obesity is
entific basis for the prevention and treatment of pedi-
associated with high levels of circulating inflamma-
atric obesity. Int J Obes. 2014;38(7):887–905.
tory mediators, and low insulin sensitivity. Int J Obes.
10. Jensen MD, Ryan DH, Apovian CM, Ard JD,

2014;38(1):46–52.
Comuzzie AG, Donato KA, Hu FB, Hubbard VS,
4. Ogden CL, Carroll MD, Kit BK, Flegal
Jakicic JM, Kushner RF, Loria CM, Millen BE, Nonas
KM. Prevalence of childhood and adult obesity in the
CA, Pi-Sunyer FX, Stevens J, Stevens VJ, Wadden
United States, 2011–2012. JAMA. 2014;311(8):
TA, Wolfe BM, Yanovski SZ, American College of
806–14.
Cardiology/American Heart Association Task Force
5. Oakley CB, Scher AI, Recober A, Peterlin
on Practice G, Obesity S. 2013 AHA/ACC/TOS
BL. Headache and obesity in the pediatric population.
guideline for the management of overweight and obe-
Curr Pain Headache Rep. 2014;18(5):416.
sity in adults: a report of the American College of
Suggested Readings 361

Cardiology/American Heart Association Task Force 24. Cockrell Skinner A, Foster EM. Systems science and
on Practice Guidelines and The Obesity Society. J Am childhood obesity: a systematic review and new direc-
Coll Cardiol. 2014;63(25 Pt B):2985–3023. tions. J Obes. 2013;2013:129193.
11. Garcia-Hermoso A, Saavedra JM, Escalante Y,
25. Cook S, Kavey RE. Dyslipidemia and pediatric obe-
Sanchez-Lopez M, Martinez-Vizcaino V. sity. Pediatr Clin North Am. 2011;58(6):1363–73. 6.
Endocrinology and adolescence: aerobic exercise 26. Kelly AS, Jacobs Jr DR, Sinaiko AR, Moran A,

reduces insulin resistance markers in obese youth: a Steffen LM, Steinberger J. Relation of circulating oxi-
meta-analysis of randomized controlled trials. Eur J dized LDL to obesity and insulin resistance in chil-
Endocrinol. 2014;171(4):R163–71. dren. Pediatr Diabetes. 2010;11(8):552–5.
12. DeBoer MD, Gurka MJ. Low sensitivity of the meta- 27. Fernandez JR, Redden DT, Pietrobelli A, Allison

bolic syndrome to identify adolescents with impaired DB. Waist circumference percentiles in nationally
glucose tolerance: an analysis of NHANES 1999-­ representative samples of African-American,
2010. Cardiovasc Diabetol. 2014;13:83. European-American, and Mexican-American chil-
13. Courcoulas AP, Yanovski SZ, Bonds D, Eggerman dren and adolescents. J Pediatr. 2004;145(4):439–44.
TL, Horlick M, Staten MA, Arterburn DE. Long-term 28. Weiss R, Lustig RH. Chapter 22 - Obesity, metabolic
outcomes of bariatric surgery: A National Institutes of syndrome, and disorders of energy balance. In:
Health Symposium. JAMA Surg. 2014;149(12): Sperling MA, editor. Pediatric endocrinology.
1323–9. 4th ed. 2014. p. 956–1014.
14. Bosch TA, Dengel DR, Kelly AS, Sinaiko AR, Moran 29. Levy-Marchal C, Arslanian S, Cutfield W, Sinaiko A,
A, Steinberger J. Visceral adipose tissue measured by Druet C, Marcovecchio ML, Chiarelli F. Insulin resis-
DXA correlates with measurement by CT and is asso- tance in children: consensus, perspective, and future
ciated with cardiometabolic risk factors in children. directions. J Clin Endocrinol Metab. 2010;95(12):
Pediatric Obesity. 2014 Jun;10(3):172–9. 5189–98.
15. Weintraub Y, Singer S, Alexander D, Hacham S,
30. Ingelfinger JR. Clinical practice. The child or adoles-
Menuchin G, Lubetzky R, Steinberg DM, Pinhas-­ cent with elevated blood pressure. N Engl J Med.
Hamiel O. Enuresis–an unattended comorbidity of 2014;370(24):2316–25.
childhood obesity. Int J Obes. 2013;37(1):75–8. 31. Inge TH, Krebs NF, Garcia VF, Skelton JA, Guice KS,
16. Sherafat-Kazemzadeh R, Yanovski SZ, Yanovski
Strauss RS, Albanese CT, Brandt ML, Hammer LD,
JA. Pharmacotherapy for childhood obesity: present Harmon CM, Kane TD, Klish WJ, Oldham KT,
and future prospects. Int J Obes. 2013;37(1):1–15. Rudolph CD, Helmrath MA, Donovan E, Daniels
17. Sbruzzi G, Eibel B, Barbiero SM, Petkowicz RO, SR. Bariatric surgery for severely overweight adoles-
Ribeiro RA, Cesa CC, Martins CC, Marobin R, cents: concerns and recommendations. Pediatrics.
Schaan CW, Souza WB, Schaan BD, Pellanda 2004;114(1):217–23.
LC. Educational interventions in childhood obesity: a 32. de Franca GV A, Restrepo-Mendez MC, Loret de
systematic review with meta-analysis of randomized Mola C, Victora CG. Size at birth and abdominal adi-
clinical trials. Prev Med. 2013;56(5):254–64. posity in adults: a systematic review and meta-­
18. Huang JS, Barlow SE, Quiros-Tejeira RE, Scheimann analysis. Obes Rev. 2014;15(2):77–91.
A, Skelton J, Suskind D, Tsai P, Uko V, Warolin JP, 33. Diesel JC, Eckhardt CL, Day NL, Brooks MM,

Xanthakos SA. Childhood obesity for pediatric gas- Arslanian SA, Bodnar LM. Is gestational weight gain
troenterologists. J Pediatr Gastroenterol Nutr. 2013; associated with offspring obesity at 36 months?
56(1):99–109. Pediatr Obes. 2015 Aug;10(4):305–10.
19. Hoelscher DM, Kirk S, Ritchie L, Cunningham-Sabo 34. Gillman MW, Ludwig DS. How early should obesity
L, Academy Positions Committee. Position of the prevention start? N Engl J Med. 2013;369(23):
Academy of Nutrition and Dietetics: interventions 2173–5.
for the prevention and treatment of pediatric over- 35. Hanson MA, Gluckman PD. Developmental origins
weight and obesity. J Acad Nutr Diet. 2013; of health and disease - Global public health impli-
113(10):1375–94. cations. Best Pract Res Clin Obstet Gynaecol.
20. Flynn J. The changing face of pediatric hypertension 2015;29(1):24–31.
in the era of the childhood obesity epidemic. Pediatr 36. van Dijk SJ, Molloy PL, Varinli H, Muhlhausler BS,
Nephrol. 2013;28(7):1059–66. members of Epi S. Epigenetics and human obesity. Int
21. D’Adamo E, Santoro N, Caprio S. Metabolic syn- J Obes. 2015;39(1):85–97.
drome in pediatrics: old concepts revised, new con- 37. Dabelea D, Crume T. Maternal environment and the
cepts discussed. Curr Probl Pediatr Adolesc Health transgenerational cycle of obesity and diabetes.
Care. 2013;43(5):114–23. Diabetes. 2011;60(7):1849–55.
22. Cote AT, Harris KC, Panagiotopoulos C, Sandor GG, 38. Expert panel on integrated guidelines for cardiovascu-
Devlin AM. Childhood obesity and cardiovascular dys- lar health and risk reduction in children and adoles-
function. J Am Coll Cardiol. 2013;62(15):1309–19. cents: summary report. Pediatrics. 2011;128 Suppl
23. Cohen AK, Rai M, Rehkopf DH, Abrams B.
5:S213–56.
Educational attainment and obesity: a systematic 39. Barlow SE. Committee at E. Expert Committee

review. Obes Rev. 2013;14(12):989–1005. Recommendations Regarding the Prevention,
362 13 Obesity

Assessment, and Treatment of Child and Adolescent Status 3 Years after Bariatric Surgery in Adolescents.
Overweight and Obesity: summary report. Pediatrics. N Engl J Med. 2015 Nov 6.
2007;120(Supplement 4):S164–92. 44. Evidence Analysis Library Academy of Nutrition and
40. Zimmet P, Alberti KG, Kaufman F, Tajima N,
Dietetics. http://www.andeal.org/search.cfm?.
Silink M, Arslanian S, Wong G, Bennett P, Shaw J, 45. Klish WJ. Definition; epidemiology; and etiology of
Caprio S. IDF Consensus Group. The metabolic obesity in children and adolescents. http://www.upto-
syndrome in children and adolescents - an IDF con- date.com/contents/definition-epidemiology-
sensus report. Pediatr Diabetes. 2007 Oct;8(5): and-etiology-of-obesity-in-children-and-adolescents.
299–306. 46. Klish WJ. Clinical evaluation of the obese child and
41. Schwimmer JB, Deutsch R, Kahen T, Lavine JE, Stanley adolescent. http://www.uptodate.com/contents/clinical-
C, Behling C. Prevalence of fatty liver in children and evaluation-of-the-obese-child-and-adolescent.
adolescents. Pediatrics. 2006;118(4):1388–93. 47. Klish WJ. Comorbidities and complications of obe-
42. Gabbert C, Donohue M, Arnold J, Schwimmer
sity in children and adolescents. http://www.uptodate.
JB. Adenovirus 36 and obesity in children and adoles- com/contents/comorbidities-and-complications-
cents. Pediatrics. 2010;126(4):721–6. of-obesity-in-children-and-adolescents.
43. Inge TH, Courcoulas AP, Jenkins TM, Michalsky MP, 48. Franks PW, Hanson RL, Knowler WC, Sievers ML,
Helmrath MA, Brandt ML, Harmon CM, Zeller MH, Bennett PH, Looker HC. Childhood obesity, other
Chen MK, Xanthakos SA, Horlick M, Buncher CR; cardiovascular risk factors, and premature death. N
Teen-LABS Consortium. Weight Loss and Health Engl J Med. 2010;362(6):485–93.
Pediatric Endocrine Emergencies
14

Much of this information is derived from the – You may assume that salt-losing adre-
previous chapters but is summarized in this for- nal insufficiency is present if sodium
mat. It is much preferable to read the indicated decreases and potassium increases in a
chapter, which covers the issues in detail. child with ambiguous genitalia and
treat appropriately while diagnostic
measures are pending. If the child is a
Acute Adrenal Insufficiency phenotypically normal male, the pro-
vider will have no physical clues to the
Patients might present with acute adrenal insuf- diagnosis in a newborn, but if a viril-
ficiency if they have undiagnosed adrenal failure ized female infant or child presents
or if they have been treated with high-dose gluco- with hyponatremia and hyperkalemia
corticoids over 10 days or more and have recently after the neonatal period the provider
been abruptly taken off this medication. Adrenal should consider adrenal insufficiency.
insufficiency may present with signs of glucocor- • Have a history of microphallus in boys
ticoid deficiency such as hypotension and hypo- (suggest hypothalamic deficiency of
glycemia or with combined glucocorticoid and gonadotropins and possibly ACTH).
mineralocorticoid deficiency which will add • Have evidence of hyperpigmentation
hyponatremia and hyperkalemia to the situation on skin, flexural surfaces, or mucous
and more likely lead to shock. membranes which would indicate ele-
vated ACTH secretion.
1. Signs and symptoms • Have a history of cessation or rapid
(a) Adrenal insufficiency should be suspected tapering of previous glucocorticoid
in children or adolescents who: maintenance or high dose therapy
• Have a history of central nervous sys- because of preexisting conditions of
tem (CNS) surgery, trauma, tumors, or the pituitary gland or adrenal gland.
congenital defects of the CNS, as noted • Have a history of cessation or rapid
in Chap. 3. tapering of previous glucocorticoid
• Have a history of ambiguous genitalia. maintenance or high dose therapy
– In this case, investigate possible viril- because of numerous conditions treated
izing or salt-losing congenital adrenal with glucocorticoid therapy including
hyperplasia, as described in Chap. 8. neoplasia, autoimmune disease, allergies,

© Springer International Publishing Switzerland 2016 363


D.M. Styne, Pediatric Endocrinology, DOI 10.1007/978-3-319-18371-8_14
364 14 Pediatric Endocrine Emergencies

pulmonary disease, and renal disease but should be obtained for future reference
among others. if adrenal insufficiency is suspected.
• Have midline defects of the head which • Adrenocorticotropic hormone (ACTH)
might be an indicator of hypo-pituitarism. may be high or low depending on etiol-
(b) The patient might exhibit symptoms of ogy from adrenal disease or hypotha-
acute adrenal insufficiency which could lamic-pituitary disease. Likewise this
be severe enough to lead to death and if will not return until well after the
mineralocorticoid deficiency also occurs, emergency situation but may be help-
severe electrolyte abnormalities will be ful in making the diagnosis.
noted. Symptoms of chronic glucocorti- • Serum Na is low and K is high if min-
coid deficiency include hypoglycemia eralocorticoids are affected, as well as
and hypotension and shock, whereas min- glucocorticoids.
eralocorticoid deficiency is heralded by • Serum/blood glucose is low.
hyponatremia with hyperkalemia, low- NOTE: Acute adrenal insufficiency
voltage electrocardiogram (EKG) with may present with hypoglycemia which,
small heart on radiograph, tachycardia, unless the child is known to have adre-
and acidosis and acidemia. nal insufficiency or glucocorticoid
Signs that may indicate acute or chronic dependence, should lead the provider
adrenal insufficiency include: to obtain a critical blood sample which
• Weakness; fatigue OR malaise; leth- will allow the diagnosis of the etiology
argy increasing to anorexia, nausea, of hypoglycemia. The critical blood
vomiting; fever; a history of weight sample is discussed in Chap. 12.
loss or failure to gain weight, history of • Low-voltage electrocardiogram (EKG)
hypoglycemia, and abdominal pain. with tachycardia and small heart on
• Dehydration with tachypnea, postural radiograph.
hypotension, decreased skin turgor, or • EKG changes due to hyperkalemia
shock. (peaked T waves).
• Petechiae and purpura may occur • Hypochloremic acidosis.
if meningococcemia or Waterson– • Eosinophilia due to decreased cortisol
Friderichsen occurs. is possible.
• Proximal and symmetrical weakness 2. Glucocorticoid replacement is accomplished by:
of the muscles due to steroid-induced (a) Immediate administration of hydrocortisone
due to prior glucocorticoid therapy. in a dose of 1–2 mg/kg or 60 mg/m2 or 50 mg
• Hypoglycemic seizures are possible. for children under 4 years of age and 100 mg
• Physical features of Cushing disease as for those over 4 years of age by subcutaneous,
described in Chap. 10 which would intramuscular, or intravenous route. No sig-
suggest long-term glucocorticoid nificant side effects are likely from giving too
exposure which might have recently much glucocorticoid replacement acutely, so
been interrupted. err on the higher dose if necessary. Cortisone
• Postural hypotension or an increase in acetate is not an emergency medication as it
pulse rate on standing may be seen; a takes too long to exert its effect as is converted
decrease of blood pressure of 10 mmHg to the active principle, hydrocortisone.
or an increase in pulse rate of 20 is (b) Stress doses are three times maintenance
considered significant. dosage (i.e., 50 mg/m2 of cortisol or
• Hypotensive or hypoglycemic shock. 7.5 mg/m2 of 6-αmethylprednisolone
• Coma or death is possible. (Solumedrol)); this is considered if the
(c) Laboratory evaluation reveals: patient is affected by infection, surgery, or
• Low-serum cortisol (cortisol should severe illness and is continued until the
increase in a stressful situation): This value situation improves or until the patient is
will not return in an emergency situation stable.
Diabetic Ketoacidosis 365

(c) The maintenance phase is accomplished ketones present venous pH less than or equal to
by the administration of standard replace- 7.30 and bicarbonate equal to 15 mEq/L or less.
ment doses of glucocorticoid (6–15 mg/ 2. Measurement of sodium, potassium, blood
m2/day orally, divided into four or three urea nitrogen (BUN), creatinine, phosphorus,
doses/day). This can be given by mouth if magnesium, and white blood cell (WBC) count
the patient is able to tolerate oral [usually elevated in diabetic ketoacidosis
administration or by i.v. or subcutaneously (DKA) even without infection] is performed.
(the oral dose is double the parenteral 3. Vital signs and neurological checks must be
dose) until maintenance therapy is started. checked frequently hourly.
3. Hypoglycemia is treated as described later. 4. Fluid and electrolyte replacement
4. Hyponatremia with hyperkalemia is treated as (a) Although excessive and rapid fluid admin-
described later. istration might cause serious complica-
5. Volume expansion is accomplished by 20 mL/ tions, dehydration leading to thrombosis
kg of 5 % dextrose and normal saline as a is an alternative risk, so each patient must
bolus in the first hour and maintenance or be individualized for appropriate fluid and
replacement fluids thereafter in accordance electrolyte therapy.
with standard fluid management. (b) Fluid administration must be done with
6. It is imperative that if hypothyroidism and glu- care, but if indicated by signs of fluid defi-
cocorticoid deficiency coexist that glucocorti- cit, an i.v. bolus of 10 mL/kg of “isotonic”
coid be administered before or with thyroid fluid (0.9 % saline or lactated Ringer’s)
hormone, or any minimal glucocorticoid that over a 30- to 60-min period is used and
may be present endogenously which may be repeated if necessary.
exerting some effect will be more rapidly metab- (c) Maintenance fluids are added to the defi-
olized, leading to acute glucocorticoid-deficient cit replacement.
crisis. Thyroid metabolism changes slowly, so (d) Potassium will be deficient in DKA, so
there is no effect of thyroxine given in the emer- after establishing urine output, add potas-
gency room, but the effects will occur in several sium to the infusion at 30–40 mEq/
days later which might be the time frame in mL. Monitor electrocardiogram (EKG)
which such adrenal insufficiency will manifest. for abnormalities of potassium, as peaked
7. Pay careful attention to the type of glucocorti- T waves occur in hyperkalemia and low
coid being used for emergency treatment. T waves and the development of U waves
Hydrocortisone sodium succinate known as occur in hypokalemia. Adequate urine
Solu-Cortef or methylprednisolone sodium output must be established before admin-
succinate known as Solu-Medrol are fast- istering potassium.
acting preparations. However, the administra- (e) Low phosphorus is a potentially danger-
tion of cortisone acetate or prednisone will ous complication of DKA, so KPO4 is
require conversion to the active moiety and used as at least a portion of the replace-
should not be used in emergency situations. ment of K if Calcium is normal.
(f) Evaluate clinical deficit, which will usu-
ally be between 5 and 12 % or even 15 %;
average is 7 % dehydration; the following
Diabetic Ketoacidosis can be altered for more significant
dehydration.
1. The diagnosis of diabetic ketoacidosis requires • Deficit: Assume 7 % dehydrated;
a blood sugar over 200 mg/dL, there is ketone- weight (kg) × 70 = (a) _____mL.
mia determined by a serum beta-OH butyrate • Remaining deficit; subtract all boluses
greater than 4 mEq/L or ketones strongly posi- given: (a) − bolus IVF = (b) _____mL.
tive by ketostix measurement at a greater than • Replace remaining deficit over 48 h:
1:2 dilution of serum, ketonuria with urinary (b)/48 h = (c) _____mL/h.
366 14 Pediatric Endocrine Emergencies

• Maintenance IVF: (d)_____mL/h. infusion may continue until the serum


• Total IVF (maintenance + remaining bicarbonate is above 17–18 mEq/mL.
deficit): (c) + (d) = _______mL/h. 6. Monitoring
(g) If serum glucose > 250 mg/dL (a) Measure blood sugar by glucometer
• Normal saline with 20 mEq KCl and or laboratory determinations every hour and
20 mEq KPO4 at total IVF rate. aim to decrease blood glucose by 10 mg/
(h) If serum glucose < 250 mg/dL dL/h. This rate may easily be exceeded, as
• D10 1/2 normal saline with 20 mEq renal function, fluid, and insulin administra-
KCl and 20 mEq KPO4 at 1/2 total IVF tion all exert effects on decreasing blood
rate plus 1/2 normal saline with 20 mEq sugar, so careful observation is mandatory.
KCl and 20 mEq KPO4 at 1/2 total IVF Check extreme values with a laboratory
rate to make D5 1/2 NS with 20 mEq determination of glucose.
KCl and 20 mEq KPO4 at total IVF (b) Measure serum electrolytes, PO4, and Mg
rate. (Note; there is controversy over every 6 h.
whether normal saline should be used (c) Measure arterial or venous blood gasses
in almost all situations including this. every 3 h.
You must use clinical judgement in the (d) Perform at least hourly neurological exams.
condition of the patient to decide on Watch for deteriorating mental condition,
this issue) headaches, vomiting, or unusual behavior, as
(i) The remaining fluid deficit is adminis- these might indicate the development of cere-
tered over the next 36–48 h. bral edema. Physical signs might include
(j) Bicarbonate therapy is associated with a changing neurologic examination and papill-
higher risk for cerebral edema and so should edema. If necessary, confirm the develop-
not be used unless symptomatic hyperkale- ment of cerebral edema with head magnetic
mia is found or the acidosis is severe enough resonance imaging (MRI), but adequate
to cause hemodynamic instability not medical support must accompany the patient
responsive to other therapeutic measures. If in case herniation becomes imminent.
pH is < 7.0 and there is cardiovascular insta- Treatment of cerebral edema is difficult, and
bility or symptomatic hyperkalemia, bicar- generally the methods are unproven, but
bonate at a dose of 1 mEq/kg over 60 min mannitol (0.25– 1 mg/kg, i.v., given over 20
may be considered to extend; this is rarely minutes every 2–4 hours or a total of 10–20 g/
invoked because the use of bicarbonate is m2) may be beneficial as long as dehydration
associated with worsening intracellular aci- is not allowed to develop after the use of such
dosis (cardiovascular and CNS) and hypoka- a hyperosmolar agent.
lemia and the development of cerebral
edema; this decision must be individualized;
this chapter does not advocate routine bicar-
bonate administration. Hypocalcemia
5. Insulin management
(a) Intravenous regular human insulin at a 1. The neonate
rate of 0.075–0.1 unit/kg/h is infused, and (A) Definition of hypocalcemia
blood sugar monitored hourly. If BS is • Although there is controversy over the
over 1,000 mg/dL with mild acidosis, a cutoffs, in term infants, serum Ca < 8.5 mg/
rate of 0.05 unit/kg/h may be used. dL and ionized Ca < 4.5 mg/dL might pre-
(b) When blood sugar decreases to 250 mg/dL, cipitate symptoms but the diagnosis of
i.v. dextrose (10 %) is added to allow con- hypocalcemia is usually serum total Ca
tinued administration of insulin to bring less than 7.0 mg/dL or ionized.
about resolution of the ketoacidosis safely; • Ca2+ less than 3.5 mg/dL.
discontinue insulin infusion only if BS can- • In premature infants, serum Ca < 7.5 mg/
not be maintained over 150 mg/dL. Insulin dL and ionized Ca < 4.4 mg/dL (or
Hypocalcemia 367

1.1 mmol/L) for birth weight > 1,500 g a total dose no more than 3 grams per dose of
and < 7 mg/dL and ionized Ca < 3.6 mg/ calcium gluconate is used. Alternatively,
dL (0.9 mmol/L) for birth 10 % calcium chloride in a dose of 20 mg/kg
weight < 1,500 g. or 0.2 mL/kg might be substituted with the
(B) Signs in the neonate may be only poor same regimen. EKG monitoring for arrhyth-
feeding, emesis, crying and apnea or mias or bradycardia is mandatory at any
more specific signs including tachycar- time i.v. calcium is administered. If an
dia, irritability, jitteriness, tetany, laryn- umbilical venous catheter is used and the tip
gospasm, and frank seizures. is inadvertently inserted in the liver, hepatic
(C) Specific conditions leading to neonatal necrosis may result from IV calcium admin-
hypocalcemia are the following: istration. A repeated dose may be adminis-
• Early hypocalcemia of infancy tran- tered after 10–15 min if there has been no
siently during the first 3 days after birth response of the symptoms and every 6 h but
is most common in infants who are pre- only if clinically indicated for continued
mature, have intrauterine growth retar- hypocalcemia. Maintenance intravenous
dation (IUGR) or small for gestational calcium is 10–50 mg of elemental calcium/
age (SGA), have sepsis, have asphyxi- kg/24 h given as 10 % i.v. calcium gluconate
ation, or are infants of diabetic mothers at a dose of 100–500 mg/kg.
(the worse the control of blood sugar, (F) Severe hypomagnesemia as etiology for
the lower the Ca in the baby). This may hypoparathyroidism is treated with IM
also occur in infants fed with cow’s injection or a 1–2-h intravenous infusion
milk-based formula. of 50 % magnesium sulfate at a dose of
• Late neonatal hypocalcemia lasts lon- 0.1–0.2 mL/kg, again with careful EKG
ger than 3 days or occurs after 3 days monitoring. The goal is to achieve a mag-
and may be due to hypoparathyroidism, nesium level of 1.5 mg/dL.
dietary or renal disease associated with 2. The older child
hyperphosphatemia, increase in dietary (A) Symptoms of hypocalcemia include the
phytates due to soy formula or the early following:
introduction of cereal, hypomagnese- • Neuromuscular signs including mus-
mia, or pseudohypoparathyroidism. cle cramping, carpopedal spasms, tet-
• Suspect hypocalcemia in neonates any, weakness, and paresthesias.
with undiagnosed seizures, a family • Cardiovascular signs including
history of hypocalcemia or various Lengthening of the QTc interval may be
autoimmune diseases, tendency to seen on the electrocardiogram (EKG) or
infections, and a defect of the great ventricular dysrhythmias, hypotension,
arteries (e.g., truncus arteriosus). and syncope.
• Look for EKG abnormalities of hypo- • The respiratory system might be affected
calcemia as described below. by laryngospasm or stridor, broncho-
(D) Mild cases might be treated by a change in spasm, respiratory distress, or apnea.
formula to a low-phosphorus formula (such • Gastrointestinal symptoms include dys-
as pm 60/40) or one with at least a 4:1 cal- phasia, vomiting, and abdominal disten-
cium-to-phosphorus ratio in addition to tion, and, in infants, feeding and crying.
oral calcium glubionate (30–50 mg/kg/day (B) The classic signs of hypocalcemia are the
in four divided doses) or calcium gluconate following:
(500 mg/kg/day in 4–6 feedings per day). • The Chvostek sign, in which tapping
(E) With severe symptoms including seizures the facial nerve below the zygomatic
and laryngospasm, intravenous (i.v.) 10 % arch causes a twitch at the angle of the
calcium gluconate (equivalent of 9.3 mg/mL mouth due to contraction of the peri-
of elemental calcium) in a dose of 100 mg/ oral muscles, a demonstration of mus-
kg or 1 mL/kg infused over 15 –20 min with cular irritability.
368 14 Pediatric Endocrine Emergencies

• The Trousseau sign, in which a blood mental calcium of 400–1200 mg/day or by


pressure cuff is inflated at least 10 % i.v. calcium gluconate at a dose of 100–
15 mmHg above the systolic blood pres- 500 mg/kg over a 24-h period for neonates
sure for 2–5 min; a positive response is and 100–200 mg/kg for infants or older
indicated by carpopedal spasms in the subjects.
arm to which the cuff was applied. 5. Prolonged therapy in hypoparathyroid states
• The peroneal sign, in which a tap on includes 1,25(OH)2 vitamin D (this form,
the peroneal nerve near the lateral tib- known as rocaltrol, eliminates the need for
ial prominence causes plantar flexion 1-hydroxylation of vitamin D that is impaired
(although tapping any nerve may lead in hypoparathyroidism), given at a dose of
to contraction of the associated mus- 20–40 ng/kg/day (0.25–0.75 μg, calculated as
cle, other contractions may be more twice daily) with, if necessary, oral calcium
difficult to demonstrate). supplementation adequate to maintain serum-
(C) EKG changes of hypocalcemia include ionized calcium.
the following: 6. In the hyperphosphatemic state due to hypo-
• Prominent U wave parathyroidism, phosphorus is restricted in the
• Narrowing of the QRS complex diet until serum phosphorus decreases to near-
• Reduced PR interval normal levels, which should occur when
• T wave flattening and inversion serum-ionized calcium approaches normal.
• Prolongation of the QTc interval 7. Serum-ionized calcium should be measured
• Prolonged ST and ST depression regularly until stability is achieved to assure
(D) Etiologies of hypocalcemia: that serum calcium remains in the normal range.
• Endocrine diseases as discussed in 8. If magnesium is low and is a possible etiology
Chap. 7. for hypoparathyroidism, magnesium sulfate
• Other medical conditions and treatment (50 % solution) may be given as 0.1–0.2 mL/
include the use of phosphate-contai- kg i.m. every 12–24 h, as needed. The goal is
ning enemas and calcium chelation to achieve a magnesium level of 1.5 mg/dL.
occurring with blood transfusions.
3. While symptoms might occur if total Ca is
less than 8.5 mg/dL, hypocalcemia is defined
as serum total Ca less than 7.0 mg/dL or ion- Hypercalcemia
ized Ca2+ less than 3.5 mg/dL.
4. For severe symptomatic hypocalcemia, oral 1. Hypercalcemia is defined as serum calcium
elemental calcium of 45–64 mg/kg/day given value above the age-related normal for the
in 4 equal doses or 10 % calcium gluconate is laboratory for age, but symptoms are usually
given as a dose of 100 to 200 mg/kg/dose or noted above 10.8–11.3 mg/dL.
1–2 ml/kg/dose over 5–20 min up to a total of Symptoms of hypercalcemia are the
10 mL, given with EKG monitoring (brady- following:
cardia and asystole are possible complica- (a) Weakness and listlessness.
tions, and infusion should stop if pulse (b) Irritability.
decreases to less than 60 beats/min or given (c) Abdominal discomfort and symptoms
extremely carefully if pulse decreases to less including nausea and vomiting, abdomi-
than 100 beats/min). During the administra- nal pain, and constipation.
tion of i.v. calcium, it is important to avoid (d) CNS symptoms including depressed con-
extravasation, which may cause severe slough- sciousness and confusion.
ing of the skin, possibly requiring plastic sur- (e) Polyuria and polydipsia.
gery. The infusion may be repeated if it is • In the neonate may manifest gastroesoph-
necessary for the relief of symptoms but is ageal reflux, lethargy, decreased weight
followed by a maintenance dose of oral ele- gain, and lack of growth in length.
Hypercalcemia 369

• In the older child: (i) Child with treatment with thiazide


– Neurological symptoms including diuretics.
lethargy, weakness, inability to con- 4. Laboratory values include the following:
centrate, and depression. (a) Serum Ca more than 10.8–11.3 mg/dL.
– Abdominal discomfort and symp- (b) If parathyroid hormone (PTH) is present
toms including nausea and vomit- in excess, serum PO4 is low.
ing, abdominal pain, constipation, 5. Treatment is as follows:
and weight loss. (a) Nonsymptomatic hypercalcemia requires
• Hypercalcemia leads to polyuria due to simple measures including increasing
an inability to concentrate the urine; hydration with, e.g., sodium chloride
dehydration and azotemia may result. infusion at twice maintenance if the
2. Signs of hypercalcemia are the following: patient can tolerate increased hydration,
(a) Band keratopathy of the medial and lat- decreasing oral calcium intake, and lim-
eral margins of the cornea (nonpurulent iting vitamin D intake including
conjunctivitis). multivitamins.
(b) EKG changes of hypercalcemia include (b) For severe hypercalcemia furosemide,
the following: 1 mg/kg, may be given by i.v. slowly if
• With mild hypercalcemia, broad-based calcium is not decreasing, to increase
tall peaking T waves urinary calcium excretion. Because the
• With severe hypercalcemia patient may initially have some degree of
– Extremely wide QRS dehydration, the sodium chloride infusion
– Low R wave must be used in concert with the diuretic.
– Disappearance of P waves Thiazide diuretics should not be used in
– Tall peaking T waves hypercalcemia if it is able to enhance
• Shortening of the QTc interval calcium retention.
• Shortening of the ST interval (c) Sodium sulfate and sodium phosphate
(c) Hypertension. may be administered and other phos-
(d) Hypercalciuria and nephrolithiasis. phates to bind calcium in the intestine, but
(e) Pancreatitis. oral phosphates may result in diarrhea.
(f) Peptic ulcer disease. (d) Oral glucocorticoids (prednisone at
(g) If serum calcium increases to more than 1–2 mg/kg/day) decrease calcium absorp-
16 mg/dL, stupor and coma may develop. tion from the intestines and may be used
3. Suspect hypercalcemia in a: in severe cases of hypervitaminosis D but
(a) Child with history of hypoparathyroidism are not effective in cases of hyperparathy-
treated with a long acting vitamin D prepa- roidism.
ration, perhaps with calcium replacement. (e) If serum calcium is greater than 14 mg/dL,
(b) Child with family history of multiple calcitonin (2–4 units/kg/injection s.c., every
endocrine neoplasia type 2 (MEN 2). 6–12 h as needed) or bisphosphonates
(c) Child with mucosal neuromas characteristic (e.g., pamidronate, 0.5–1.0 mg/kg/dose i.v.
of multiple endocrine neoplasia type 3 over 4–5 h, repeated once as necessary up
(MEN 3). to 2 mg/kg/dose) may be effective, but nei-
(d) Immobilized child. ther are FDA approved for children.
(e) Child with various malignancies including 6. If renal failure coexists, renal dialysis might
leukemia. be necessary to decrease serum calcium.
(f) Child with megavitamin therapy particu- 7. If malignancy-derived PTH-related protein
larly with vitamin D or A. (PTHRP) is the etiology, Plicamycin (for-
(g) Child with physical features of Williams merly known as mithramycin) at 25 μg/kg/day
syndrome. may be used but may not be available; liver
(h) Neonate with subcutaneous fat necrosis. toxicity is a side effect.
370 14 Pediatric Endocrine Emergencies

Hyperthyroidism in the Neonate 4. Emergency treatment includes the following:


(a) Propranolol (1–2 mg/kg/day divided into
1. Hyperthyroidism is rare as a primary diagnosis three or four oral doses) may be used to
in the newborn but is seen in infants whose counter the sympathetic effects of hyper-
mothers have hyperthyroidism (which might thyroidism (see later).
yet not be diagnosed in the mother). If the (b) Corticosteroids given as an intravenous
mother has previously diagnosed hyperthyroid- injection of 5 mg/kg of hydrocortisone
ism and was taking oral meds, e.g., imidazole or (the maximal dose of 100 mg ) inhibit the
propylthiouracil to treat the hyperthyroidism, conversion of thyroxine to T3 and
the medication will pass through the placenta to decrease secretion of thyroxine and the
the fetus and the newborn may have a positive thyroid gland.
hypothyroid screen initially only to develop (c) Lugol’s solution of supersaturated potas-
symptoms of hyperthyroidism several days sium iodine may be used to suppress thy-
after birth when the antithyroid medication is roid hormone secretion (one drop every
metabolized and eliminated. Sometimes the 8 h is usually adequate).
appearance of hyperthyroidism in this situation (d) Some have suggested the use of digitalis
occurs weeks after birth. or sedatives might be indicated in severely
affected neonates.
If the mother is receiving excessive doses of (e) A severely affected neonate with hyper-
antithyroid medication, the fetus may develop a thyroidism may have symptoms for up to
goiter so severe that respiratory compromise 6 weeks or more and for that reason
occurs immediately after birth: newborns have methimazole is started (0.25–1 mg/kg/
died in this situation. day divided into three doses). However,
the child must be watched closely for res-
2. Signs of hyperthyroidism in the newborn olution of hyperthyroidism that usually
include the following: occurs before 12 weeks or sooner.
(a) Neurologic symptoms including shakiness. Continued treatment with methimazole
(b) Cardiovascular signs including tachycar- when thyroid function escapes from stim-
dia or even congestive heart failure due to ulation by the maternal immunoglobulins
supraventricular tachycardia. (Some would lead to iatrogenic hypothyroidism
babies have been admitted to the neonatal and subsequent developmental delay.
intensive care unit with supraventricular
tachycardia and may receive suboptimal
treatment if hyperthyroidism is not ini- Thyroid Storm in Children
tially suspected.) and Adolescents
(c) Hepatosplenomegaly may occur with the
development of congestive heart failure. 1. This is a rare complication to be suspected in
3. Laboratory findings include: children with:
(a) Elevated serum-free T4 or total T4 accord- (a) A history of hyperthyroidism with
ing to age-appropriate standards for intercurrent
newborns according to the laboratory per- • Infection
forming the analysis. • Surgery including thyroid surgery
(b) TSH is usually suppressed as the thyroid • Radioactive iodine therapy
gland is stimulated by immunoglobulins • Diabetic ketoacidosis
in this condition. As stated above, TSH • Pregnancy
might be elevated in the first days after (b) Alternatively it can occur as the first
birth in infancy if the mother was taking manifestation of an undiagnosed case of
thyroid-suppressive medication. hyperthyroidism.
Hypoglycemia 371

2. Thyroid storm is indicated by an acute onset (f) Appropriate fluid management must be
of hyperthermia and tachycardia in a patient instituted, and if tachycardia causes heart
with underlying hyperthyroidism. It may failure, digitalis may be necessary.
occur during surgical or radioiodine therapy
for hyperthyroidism.
3. Symptoms include Hypoglycemia
(a) High fever
(b) Sweating 1. Diagnosis is made if:
(c) Tachycardia (a) Plasma glucose is less than 60 mg/dL or,
(d) Reduced mental status, ranging from con- because whole-blood glucose is 15 % lower
fusion to coma than serum or plasma glucose, blood glu-
4. Immediate therapy is indicated. cose is less than 50 mg/dL or capillary sugar
(a) Propranolol is used in a starting dose of is less than 60 mg/dL (but not when the
1–3 mg/kg/day, divided into an oral dose blood has been sitting for hours at room
every 6 h to control adrenergic symptoms temperature allowing the red cells to metab-
of thyroid storm. Propranolol may be given olize the glucose bringing down the value).
i.v. at a dose of 0.01 starting dose mg/kg, (b) A critical sample must be obtained if BS is
up to a total of 5 mg over 10–15 min, but <50 mg/dL or an important opportunity to
an intraatrial pacing catheter is a necessary make a diagnosis of the etiology of hypo-
precaution: start with the lower range of glycemia will be lost. Obtain, if safe,:
doses. Atenolol is suggested to be prefera- • Glucose
ble to propranolol due to its more selective • Insulin
cardiac effects but there is little experience • Ketones
in children. It is reported to be used in 1–2 • Growth hormone
mg/kg per day as a single daily dose or • Cortisol
given every 12 hours; maximum is reported • Lactate
to be 100 mg/dose. • Pyruvate
(b) Dexamethasone in a dose of 1–2 mg (i.v. And if possible:
dose is 0.2 mg/kg) and/or hydrocortisone • Free fatty acids
at 5 mg/kg every 6 h (the maximal dose of • C-peptide
100 mg ) can decrease conversion of T4 to • IGF-1
T3 and decreases secretion of thyroxine • IGF-11
from the thyroid gland. • IGF BP1
(c) Intravenous NaI in a dose of 125–250 mg • Ammonia
up to 1–2 g/day may decrease the release • Amino acids
of thyroid hormone from the thyroid • Free and bound carnitine
gland; Lugol’s solution of concentrated • Save as much serum/plasma as possi-
iodine in a dose of five drops once every ble for future determinations
8 h by mouth should be given if the (c) Hypoglycemia is suspected in a child with a
patient is conscious. host of resulting conditions including sepsis
(d) A cooling blanket or tepid bath can help or exposure to toxins. In terms of endocrine
control the hyperpyrexia as can acetamin- disorders, the following is a basic list:
ophen (do not use aspirin in childhood). • Child with diabetes mellitus receiving
(e) Methimazole will not take effect for sev- insulin treatment
eral days, but to plan for the likely • Child with disorder of carbohydrate
extended course of the disorder, a dose of metabolism
0.6–0.7 mg/kg/d up to 20–30 mg/day can • Child with congenital hypopituitarism
be given every 6 h by slurry, if necessary. • Boy with microphallus
372 14 Pediatric Endocrine Emergencies

• Child with midline defect honey (only if the child is well older than 1
• Child recently discontinued on sup- year and not at risk for infant botulism), or
pressive doses of glucocorticoids glucose gel/jelly can be ingested or admin-
• Child fasted for extended period istered to the child’s buccal mucosa if the
• Infant who was recently exposed to child is too disoriented to swallow.
fructose and sucrose and developed (b) For more severe hypoglycemia, i.v. glu-
symptoms thereafter cose is necessary.
2. Presenting symptoms awakening concern for • Dextrose in water is given in 25 % con-
hypoglycemia in the newborn are nonspecific centration or less as a bolus of 1–2 mL/
and range from: kg or 0.25–0.5 g of dextrose/kg body
(a) Grand mal or localized seizures to weight: alternatively, 1–2 mL/kg/dose
(b) Irritability of 10 % dextrose; 0.1–0.2 mg/kg/dose
(c) Hypotonia may also be used for initial therapy. If
(d) Lethargy more dextrose in higher concentration
(e) Difficulty in feeding, to (dextrose vials are supplied as 50 %) is
(f) Other rather nonspecific findings, to administered, a risk exists of causing
(g) No findings at all because of the immatu- hyperosmolality, and in infants and neo-
rity of the CNS nates, D10W is preferable to any more
(h) Hypoglycemia may occur during sepsis concentrated preparation.
and some of the hypoglycemia symptoms • The dextrose percentage may be multi-
may be mistaken for sepsis plied by the number of milliliters
3. Presenting symptoms in the child are administered per kilo and the result
(a) Adrenergic including should equal 50 or less for appropriate
• Sweating therapy.
• Shaking • Glucagon Emergency Kit for children
(b) Neuroglycopenic with diabetes or hyperinsulinism if i.v.
• Confusion access proved difficult. Glucagon will
• Stupor not work in most types of ketotic
• Coma hypoglycemia.
• Seizures • Once the blood sugar increases, the
4. Physical exam aims to rule out child should be observed for improved
(a) Hyperpigmentation of primary adrenal clinical condition; if the sugar again
insufficiency. decreases, an infusion of dextrose,
(b) Microphallus in a boy. 5–10 %, should be started. Sodium
(c) Midline defects, optic nerve hypoplasia, chloride must be in the maintenance
and pendular nystagmus, all suggestive of dextrose infusion to avoid hyponatre-
hypopituitarism. mia developing as the hypoglycemia is
(d) Hepatomegaly which might be found in repaired. The infusion rate will usually
glycogen storage disease type 1, gluconeo- be 6–8 mg/kg/min of glucose which is
genesis defects, hereditary fructose intoler- approximately 1.5 times maintenance.
ance, or fatty accident oxidation disorders. • If a glucose infusion rate of more than
5. The initial treatment of hypoglycemia must be 12–15 mg/kg/min of glucose is
the administration of glucose sufficient to required or repeated larger glucose
bring the blood glucose to adequate levels to boluses needed, the child has severe
return neurologic function to normal but not hypoglycemia, most likely due to
excessive glucose which will cause severe hyperinsulinism. Abrupt discontinua-
increased osmolality. tion of dextrose support may lead to
(a) In a child who is only moderately symptom- rebound hypoglycemia in a child who
atic, oral glucose is adequate; table sugar, does not have hyperinsulinism due to
Hyponatremia/Hyperkalemia Due to Adrenal Insufficiency 373

the normal increase in insulin secre- generalized: follow the guidelines for hypo-
tion caused by dextrose administra- glycemia listed earlier.
tion. This rebound condition should 4. Hyponatremia is treated as follows:
not be confused with bona fide (a) With the intravenous infusion of normal
hyperinsulinism. saline in normal replacement volumes.
• As transient hypoglycemia resolves, (b) If the patient is seizing from hyponatre-
weaning off i.v. dextrose is followed by mia, 3 % NaCl might be infused in doses
frequent oral administration of glucose- sufficient to increase the serum Na until
containing substances as the child can seizures stop; 3 mL/kg every 10–20 min is
tolerate under careful observation. a useful dose.
• Prolonged treatment aims to achieve a 5. Hyperkalemia
value of more than 60 mg/dL at all (a) Replacement of i.v. fluid with sodium will
times and normal values of 70–120 mg/ usually reduce potassium levels to a
dL at least most of the time. degree or may normalize them.
(b) If the EKG demonstrates the following
abnormalities, calcium infusion may be
Hyponatremia/Hyperkalemia administered but is quite dangerous (as is
Due to Adrenal Insufficiency severe hyperkalemia):
• Peaked T waves
1. Laboratory values include • Widening of the PR interval
(a) Serum sodium less than 135 mg/dL. • First-degree heart block
(b) Serum potassium greater than the upper • Loss of the P wave
limits of normal for age the laboratory • Ventricular arrhythmia
has used. For example: (c) 10 % calcium gluconate at a dose of
• Above 7.2 mEq/L in premature infants 0.5 mL/kg i.v., over 5–20 min, may be
• Above 5.2 mEq/L in term newborns administered to stabilize the membranes.
• Above 5.8 mEq/L in older children • If pulse decreases to less than 60 beats/
• Above 5.5 mEq/L in adults min, calcium infusion should cease.
2. This situation is suspected in • If the pulse decreases to less than
(a) Children with ambiguous genitalia who 100 beats/min, calcium infusion can be
might have virilizing congenital adrenal continued, but only with exceptional
hyperplasia. care under continued EKG monitoring.
(b) Children with a history of glucocorticoid (d) Sodium bicarbonate, 7.5 %, given as
deficiency who may also have mineralo- 2–3 mL/kg over 30–60 min, will tend to
corticoid deficiency. drive K into the cells as pH rises, reducing
(c) Children in shock (who might have adre- serum K.
nal failure). (e) 50 % Glucose plus 1 unit of insulin for
(d) Children with acquired hyperpigmentation every 5–6 g of glucose given over 30 min
who might have elevated ACTH secretion. will tend to drive K into the cells, reduc-
(e) Children with an autoimmune disease ing serum K. However, because children
such as Hashimoto’s thyroiditis who pres- with adrenal insufficiency have a ten-
ent with hyponatremia. dency toward hypoglycemia, this therapy
(f) Diabetes mellitus can present with hypo- carries danger and must be used with the
natremia due to elevated glucose or utmost caution.
osmotic diuresis in DKA but there should (f) Sodium polystyrene sulfonate
be some historical or laboratory indica- (Kayexalate) resin in a dose of 1 g/kg
tions of the presence of diabetes. (possibly given in 10 % glucose at
3. Hypoglycemia may occur as well as hypona- 1 g/4 mL) may be given every 4–6 h to
tremia because adrenal insufficiency may be increase loss of potassium in the gastroin-
374 14 Pediatric Endocrine Emergencies

testinal (GI) tract. This is not an emergency therapy. Sodium loss in the urine will continue
treatment. until SIADH is resolved, so 3 % sodium chlo-
(g) Oral Florinef is the best treatment for ride treatment used to resolve hyponatremic
hyponatremia and hyperkalemia of adre- seizures will be effective only temporarily
nal insufficiency but will take a matter of until the cause of SIADH is resolved.
hours to work. At present, no parenteral 4. Furosemide (1 mg/kg) might be used and the
treatment exists for mineralocorticoid volume excreted replaced with 3 % NaCl to
deficiency. allow an effective increase of serum sodium to
control the seizure; frank dehydration is a pos-
sible outcome if fluid volume is not watched
Hyponatremia Resulting carefully. This process is dangerous.
from Syndrome of Inappropriate 5. Dilantin, 5–10 mg/kg, has been used in patients
Secretion of Antidiuretic Hormone with seizures due to CNS etiologies of SIADH.
6. Demeclocycline is a tetracycline derivative that
1. Syndrome of inappropriate secretion of antid- reduces the ability of the nephron to respond to
iuretic hormone (SIADH) should be suspected vasopressin (AVP) and may be used for chronic
in a child who: SIADH in a dose of 10 mg/kg; this molecule is
(a) Has had CNS trauma or infection. a modification of tetracycline, this might
(b) Has increased intrathoracic pressure due adversely affect tooth development if used for
to, for example, pneumonia or is receiv- long periods in young children.
ing positive-pressure ventilation. 7. For chronic cases of SIADH, refer to Chap. 3.
(c) Has decreased serum sodium (sodium
less than 135 mg/dL), or decreased serum
osmolality in spite of:
• Urinary concentration out of propor- Hypernatremia Due to Diabetes
tion for the serum osmolality. Insipidus
• No hyperlipidemia, hyperproteinemia,
or hyperglycemia as a cause of low- 1. Diabetes insipidus is suggested in a child
serum sodium. who has:
• No concomitant hyperkalemia along (a) Increased serum osmolality
with hyponatremia. (b) Increased serum sodium
2. During the episode of hyponatremia, i.v. infu- (c) Low urine osmolality
sion of normal saline is used in a milliliter- (d) Voluminous urinary output in spite of
per-milliliter replacement given every 1–4 h high-serum osmolality
for all urinary losses; this will allow an effec- 2. This condition is suspected in a patient who
tive increase in serum sodium without an presents with:
increase in fluid volume since insensible (a) Signs of dehydration including sunken
losses will not be covered. Alternatively, with- eyes, dry mucous membranes, and sunken
holding fluids under close observation until fontanel in infants (because a tendency
the condition resolves may be tried in less exists toward hypernatremic dehydration,
severe cases. This process must consider the the loss of skin turgor may not occur, and
state of hydration so that the patient does not the skin might feel “doughy” as a result).
become clinically dehydrated and/or go into (b) Has a history of CNS tumor, surgery or
hypotensive shock while fluids are limited. accident, or bitemporal hemianopsia.
3. If the patient is seizing from hyponatremia, (c) Has a condition causing increased intra-
3 % NaCl might be infused in doses sufficient cranial pressure.
to increase the serum Na until seizures stop; (d) Has midline defects of the head which
3 mL/kg given every 10–20 min until symptoms might be indicators of congenital hypo-
of hyponatremia resolve is one recommended thalamic–pituitary disease.
Suggested Reading 375

3. This form of hypernatremia is due to loss of free 5. Patients developing hypertension during the
water rather than a condition of excess sodium. induction of anesthesia, in patients suspected
4. The length of time since dehydration began of having MEN 2 or 3, and with malignant
is important, as a chronic state will require hypertension unresponsive to usual methods
slower fluid-replacement program than in of therapy might be treated as follows:
acute cases. (a) Alpha-adrenergic block should be admin-
5. If serum sodium is greater than 160 mEq/L, a istered before β-adrenergic blockade.
bolus of normal saline in a dose of 20 mL/kg • Phenoxybenzamine is given orally (0.2
may be helpful; it is essential to bring the mg/kg/day increasing by 0.2 mg/kg/day
serum sodium down slowly. in 4 days and repeatedly until a maxi-
6. Volume deficit is calculated, and fluid replace- mum dose of 1.2mg/kg/day is reached
ment is planned for the next 48 h. and high blood pressure is controlled;
7. The volume of urine output will change doses start as once per day but split
remarkably from the large volumes before the daily doses given every 8–12 hours is
administration of DDAVP to lower volumes an alternatively) for long-term therapy.
afterward; using characteristic maintenance • Phentolamine administered i.v. or i.m.
formulas for IV fluids will lead to wide shifts (high blood pressure is controlled) is
in serum osmolality and sodium concentra- used for hypertensive crises that may
tions. Thus it is best to replace the urine vol- occur while the phenoxybenzamine
ume lost over 1–4 h with an equivalent volume block is being established. Phentolamine
of i.v. fluid over the next 1–4 h rather than is not available in the US at the time of
using simple maintenance formula to deter- this writing so an alternative Alpha
mine i.v. fluid administration. Once the vol- blocking agent must be used.
ume of urine is controlled, routine maintenance (b) β-Blockade by:
fluid calculations may again be appropriate. • Propranolol (5–10 mg given 3–4 times
8. DDAVP treatment is described as in Chap. 3, per day orally for larger children) is
but if the child cannot take oral or nasal added when the heart rate increases as
DDAVP, i.v. aqueous pitressin, at a dose of the α-adrenergic blockade is being
2–3 mg/kg/min, or i.m. administration at established.
1–5 units per dose is an alternative. • Propranolol may cause paradoxic
9. At all times, evaluation of changes in serum hypertension if given before the
sodium and urine osmolality as well as con- α-adrenergic block is established.
tinuous evaluation of the fluid balance with • If propranolol is ineffective in control-
respect to fluids administered and urinary ling blood pressure, α-methyltyrosine
excretion are essential. (a tyrosine hydroxylase inhibitor) can
be started at 250–500 mg increased to
4 grams per day given in divided dos-
Hypertension ing four times per day.
Due to Pheochromocytoma (c) Nitroprusside is used as a vasodilator
during surgical hypertension.
1. These rare tumors are described in Chap. 10. (d) Surgical preparation also includes admin-
2. Symptoms may include headaches of a pound- istration of salt to repair the plasma
ing nature, palpitations with tachycardia and volume, which is invariably low.
sweating, and possibly flushing.
3. Signs include hypertension, usually of a
constant pattern, but episodic hypertension as Suggested Reading
found in adults may occur in children.
4. Nervousness, tremor, fatigue, and thoracic Protocols are taken from the other chapters in the book,
pain may be present. with specific sources listed.
Medications for Pediatric
Endocrinology 15

The following is a list of the some of the most Calcitriol


commonly used mediations for pediatric endo- (1,25-Dihydroxycholecalciferol)
crinology. It is incumbent upon the prescriber to
check for allergies, contraindications, side Hypoparathyroidism:
effects, and safety in pregnancy. Also note that 0.04–0.08 μg/kg under 1 year of age, 0.25–0.50
many of the medications are used in clinical between 1 and 5 years and 0.05 to 2 μg/day over 6
studies and presented in publications dealing years of age.
with children without FDA approval: they are Hypocalcemia:
presented here for information, but the pre- 0.25 μg/dose, p.o./day initially. Increase by
scriber must take the age of the patient into con- 0.25 μg every 2–4 weeks while observing effects
sideration and understand that many medications on serum calcium.
below are used “off-label” and precautions and Younger than 1 year, 0.04–0.08 μg/kg/dose/ day.
consent of parents are the responsibility of the 1–5 years, 0.25–0.75 μg/dose/day.
treating physician. Older than 6 years, 0.5–2 μg/dose/day.
Some medications requiring more discussion
or used for specific testing are found in detail in
other chapters rather than listed here. Every effort Calcium
was made to check for accuracy of dosage listed
but recheking based upon the clinical situation Calcium Carbonate, Oral
and age of the child is incumbent upon the reader
who may prescribe a medication. 40 % elemental calcium, so each mg contains
0.4 mg of elemental calcium.
200mg of elemental calcium is found in 500
Calcitonin, Human mg tablet of Tums.
For calcium deficiency or for prophylaxis,
While bisphosphonates are usually used in RDA values are:
osteogenesis imperfecta, calcitonin in a dose of 2 0–6 months, 200 mg elemental calcium/day to
international units/kg three times a week with oral a maximum of 1,000 mg/day.
calcium supplements (230–345 mg/day) for up to 6–12 months, 260 mg elemental calcium per
48 months have been shown to be effective as well. day to a maximum of 1,500 mg/day.

© Springer International Publishing Switzerland 2016 377


D.M. Styne, Pediatric Endocrinology, DOI 10.1007/978-3-319-18371-8_15
378 15 Medications for Pediatric Endocrinology

1–3 years, 700 mg elemental calcium per day 1.5 mL/min or 200 mg/min. For severe
to a maximum of 2,500 mg/day. symptomatic hypocalcemia 100 to 200 mg/kg/
4–8 years, 1,000 mg elemental calcium per day dose which can be repeated if needed in 6 hours.
to a maximum of 2,500 mg/day. Alternatively 200 to 800 mg/kg/day can be given
9–13 years, 1,300 mg elemental calcium per y by continuous IV infusion.
to a maximum of 3,000 mg/day. Children: 200 up to 500 mg/kg/day which is
14–18 years, 1,300 mg elemental calcium per 2–5 mL of a 10 % solution by slow IV injection
day to a maximum of 3,000 mg/day. at about 1.5 mL/min infusion over 15–20 min-
These doses are usually divided into three utes; dose can be divided into 4–6 doses over the
portions per day. day to no more than 2–3 gram for each dose.
Alternatively a continuous infusion of 200–
800 mg/min may be given.
Calcium Citrate, Oral Oral: 400–800 mg/kg/day, divided into q.i.d.
Child: Oral or IV is 200–500 mg/kg/day,
21 % elemental calcium so that each mg of cal- divided into q.i.d.
cium citrate contains 0.211 mg elemental cal- Adult: Oral or IV is 5–15 g/day, divided into
cium. Dosing is as for calcium carbonate. q.i.d.

Calcium Glubionate, Oral Calcium Lactate, Oral

Elemental calcium (6 %), available as 115 mg 13 % elemental calcium.


elemental calcium/5 mL in a 10% solution. Infants: 400–500 mg/kg/day, divided into q4
Neonate with hypocalcemia: 1,200 mg/kg/ to 6 h.
day, divided into q.i.d. to t.i.d. Children: 500 mg/kg/day, divided into q4 to 6 h.
Maintenance: 600–2,000 mg/kg/day, up to Adults: 1.5–3 g/given t.i.d.
9 g/day, divided q.i.d.
Adults: 6–18 g/day, divided into q.i.d.
DDAVP

Calcium Gluconate IV 1-Deamino-8-D-arginine vasopressin.


Dosages of spray dispenser is 0.1 mL (10 μg)
9 % elemental calcium so that each mg of cal- per spray or 10 μg per dose.
cium gluconate contains 0.093 mg of elemental Concentration of the preparation using the
calcium; injection is 10 % solution so that each rhinal tube is 0.01 mg/0.1 mL the tube divided
mL of a 10 % calcium gluconate solution con- into 0.2, 0.15, 0.1 and 0.05 mL for 20, 15, 10 and
tains 9.3 mg (0.465 mEq) of elemental calcium. 5 μg doses.
All calcium infusion can cause severe scarring if Tablets are available in 0.1, 0.2 mg forms.
extravasation occurs. Injectable dose is 1/10 the intranasal dose.
For diabetes insipidus.
Children: 0.05 mg/dose orally given up to
Hypocalcemia 0.8 mg given qd to b.i.d.; 10–30 μg/day given
intranasally divided into qd to b.i.d.
Neonates: By slow IV injection at 100 mg/kg/day
(1 ml/kg/day) infusion over 15–20 minutes with
constant EKG monitoring; dose can be repeated Ergocalciferol (Vitamin D2)
in 10 minutes if necessary or can be divided into
4–6 doses over the day to no more than 1 gram Usually given with calcium supplementation.
for each dose. Maximum rate of infusion is Monitoring of vitamin D levels is necessary.
Hydrochlorothiazide 379

Dietary Supplementation Estrogen

Newborn infant: 400 IU/day. See Chap. 9.


Premature infants: Doses equal to or greater
than 400 IU/day with monitoring.
Infants over 1 year and children: 600 IU/day. Fludrocortisone Acetate (Florinef)

0.05–0.1 up to 0.2 mg/day, with added salt to


Vitamin D Deficiency diet, depending on the degree of salt-losing ten-
dencies and response. Watch for hypertension as
There are various schema of therapy depending an indication of excessive dosage.
upon the degree of deficiency which differ but for
example: 1,000 IU daily for infants less than 1
month for 6 weeks. Glucagon HCl
1,000–2,000 IU daily for age 1–12 months for
6 weeks. Given IV, i.m., sc or p.o. for severe hypoglycemia
2,000 IU daily for >1-year-olds or 50,000 IU if oral administration of glucose is difficult or
per week for 6 weeks. dangerous due to decreased mentation.
Monitoring 25 OH vitamin D is necessary to Neonates and children less than 20 kg: Give
avoid overdosage. 0.5 mg/dose or 0.02–0.03 mg/kg dose.
Children more than 20 kg: 1 mg/dose, repeat
in 15–20 min if necessary.
Nutritional Rickets Give oral carbohydrates if they are awake if
possible instead.
1,000–2,000 IU/day orally. Mini glucagon dose may be used to raise
200,000 IU every 3 months is an alternative blood sugar in nonemergent situations if the
method. ketones are less than moderate or large:
If there is malabsorption or hepatobiliary dis- For a child 2 years old or under, give 20 μg
ease: 10,000–25,000 IU/day. and give 10 μg more for each year of age for chil-
If there is anticonvulsant-related rickets: dren over age 2 to a maximum of 150 μg.
1,000 IU/day.

Vitamin D-Resistant Rickets Glucocorticoid Preparations

25,000–100,000 IU/day and increase as necessary. See Table 15.1 and Chaps. 8 and 10.

Vitamin D-Dependent Rickets


Growth Hormone Secretagogue
3,000–50,000 IU/day and increase as necessary. Testing

L-Dopa: 125 mg for body weight up to 15 kg,


Hypoparathyroidism or 250 mg for weight up to 35 kg, and 500 mg for
Pseudohypoparathyroidism body weight over 35 kg.
Clonidine 0.1–0.15 mg/m2.
Initiate with doses up to 8,000 IU/day for 1–2 weeks. Intravenous arginine infusion of 0.5 g/kg body
Children: 2,000 IU/day maintenance. weight up to 20 g over a 20-min period.
380 15 Medications for Pediatric Endocrinology

Table 15.1 Glucocorticoid preparations


Glucocorticoid effect
(based on anti- Mineralocorticoid effect
inflammatory effect [fludrocortisone Time of biologic
with cortisol = 1.0) (fluorine) = 1.0] action (h)
Cortisol (hydrocortisone) 1 0.005 8
Cortisone 0.8 0.005 8
Prednisone 4 0 18
Prednisolone 4–5 0 16–36
6α-Methylprednisolone 5–8 0 16–36 est
Dexamethasone 27–66 0 36–54
9α-Fluorocortisone 15 1 24–36

Insulin-induced hypoglycemia (0.075–0.1 units/ Table 15.2 Actions of commercially available insulin
kg body weight of insulin given IV) is an extremely preparations
dangerous test; read warnings in Chap. 5. Onset of Peak Duration
Glucagon 30 μg/kg up to 1 mg Preparation action activity of action (h)
Aspart 10– 40–50 min 3(to 5)
20 min
Lispro 15– 30–90 min 3(to 5)
30 min
Hydrochlorothiazide Apidra 20– 30–90 min 1–2.5
30 min
Central diabetes insipidus alternative therapy to Regular 30– 2–5 h 5–8
DDAVP. 60 min
Neonates: Younger than 6 months, 1–2 mg/ NPH 1–2 h 4–12 h 18–24
kg/day, divided b.i.d. or t.i.d., p.o. (maximum Glargine 1h None 24
dose, 37.5 mg/day). (Chlorothiazide may be used Detemir 1–2 h 6–8 h 24
instead at a dose of 5–10 mg/kg/day.) NPH neutral protamine Hagedorn

Insulin Preparations Older than 12 years: 2–3 μg/kg/dose/day


After cessation of growth dose: 1.7 μg/kg/
See Table 15.2 and Chap. 11. dose/day

Thyroxine (Levo)
Magnesium Sulfate
Note if profoundly hypothyroid, slow increase to
recommended dose is suggested to avoid poten- 1 g magnesium sulfate equals 98.6 mg, 8.12 mEq,
tial cardiac failure. or 4.06 mmol elemental Mg.
If adrenal insufficiency is suspected, glucocor-
ticoid replacement is recommended before thy-
roxine to avoid acute adrenal failure. Hypomagnesemia or Hypocalcemia
Children: p.o. dosing:
0–3 months: 10–15 μg/kg/dose/day IV (slow given over 15–20 minutes)/i.m.:
3–6 months: 6–10 μg/kg/dose/day 25–50 mg/kg/dose, divided into t.i.d. to q.i.d.;
6–12 months: 6–8 μg/kg/dose/day repeat as needed.
1–5 years: 5–6 μg/kg/dose/day Maximum single dose: 2 g.
6–12 years: 4–5 μg/kg/dose/day Oral: 100–200 mg/kg/dose, q.i.d., p.o.
Phosphate Supplements 381

Daily Maintenance Hypoglycemia is a risk of therapy, and a snack


or meal must accompany the dose within 20 min
In TPN 0.25–1.25 g magnesium sulfate (25– or the dose must be omitted. If hypoglycemia
125 mg elemental magnesium) per day for persists, the dose must be decreased.
infants.

Methimazole
Mannitol (5, 10, 15, 20, 25 %)
Hyperthyroidism
Cerebral edema: 0.25–1 g/kg/dose, IV, over
20–30 min. May increase gradually to 1–2 g/kg/ Children:
dose if needed. (May give furosemide, 1 mg/kg Initial: 0.4 mg/kg/24 h p.o., divided into q8h.
concurrently or 5 min before mannitol.) Maintenance: Half to two-thirds of initial
Maximum dose: 12.5 g over 3–5 min. dose, p.o., q8h.
However, dosage in children under 12 years is Maximum dose: 30 mg/24 h.
not officially established. Adults:
Initial: 15–60 mg/24 h from mild to severe
hyperthyroidism, p.o., divided into t.i.d.
Medroxyprogesterone Acetate Maintenance: 5–15 mg/24 h, p.o., divided
into t.i.d.
Amenorrhea: 5–10 mg, p.o., qd × 5–10 days. The total daily dose may be effective if
Abnormal uterine bleeding: 5–10 mg, p.o., divided into twice per day administration or
qd × 5–10 days, initiated on day 16 or 21 of the even one dose per day in some.
menstrual cycle.

Nitroprusside
Metformin
Children and adults: IV, continuous infusion.
Start with 500 mg p.o. with morning meal, after 1 Dose: Start at 0.3–0.5 μg/kg/min, titrate to
week if tolerated increase to 500 mg b.i.d., with effect.
morning and evening meals; may increase by Usual dose: 3 μg/kg/min.
500 mg every week, administered in divided Maximum dose: 10 μg/kg/min.
doses up to a maximum of 2,500 mg/24 h
(2,000 mg/24 h in younger teenagers).
Start with one tablet to evaluate abdominal Pamidronate
distress and then increase as needed.
Extended release forms are available. Hypercalcemia
Daily multivitamins should accompany this
treatment to avoid vitamin B12 deficiency. Children:
Mild hypercalcemia: 0.5–1 mg/kg/dose, IV ×
over 24 h. Maximal dose is 90 mg.
Mecasermin (rhIGF-1) Severe hypercalcemia: 1.5–2 mg/kg/dose, IV
× over 24 h. Maximal dose is 90 mg.
0.04–0.08 mg/kg/dose SC bid, may increase over Adults:
weeks by 0.04 mg/kg/dose to maximum of Corrected serum Ca2+: 12–13.5 mg/dL: 60 mg,
0.12 mg/kg/dose given twice daily. IV, × 1 over 4 h OR 90 mg IV, × 1 over 24 h.
382 15 Medications for Pediatric Endocrinology

Corrected serum Ca2+: More than 13.5 mg/ Thyrotoxicosis: Children: About 1–5 drops of
dL: 90 mg, IV × 1 over 24 h. SSKI containing 1 g/mL every 6 h after starting
(Dose may be repeated after 7 days.) methimazole and only temporarily.

Osteogenesis Imperfecta Potassium Supplements

Children: Doses start low on day 1 and are higher Hypokalemia


on the 2nd and 3rd day of the first 3 day cycle and
in subsequent cycles can start in the higher range. Oral:
0.25–1 mg/kg/dose, IV, may be repeated in 3–4 Children: 4–5 mEq/kg/24 h divided into b.i.d.
months for total yearly dose of 4 mg/kg/year for to q.i.d. Monitor serum potassium.
3 years. Intravenous: 0.5–1 mEq/kg/day with EKG
Maintenance is then 1 mg/kg/dose twice per monitoring and physician in attendance if higher
year for a yearly dose of 2 mg/kg/year until rate used.
growth is completed.
Monitor for hypocalcemia and hyposphatemia
before administration and thereafter. Flu like Monitor Serum K Closely and Watch
symptoms may occur at first use. EKG at All Times
Note: therapy is switching to Zoledronate for
Osteogenesis Imperfecta although this medica- Children: 0.3 mEq/kg/h, given as an infusion of
tion also is not approved for this condition. less than 40 mEq/L at a rate of no more than
20 mEq/h for 1–2 h.
This may be used in critical situations such as
Phosphate Supplements hypokalemia with arrhythmia.
Maximum peripheral IV concentration:
Note: 1 mmol phosphate= 31 mg Usually 40 mEq/L. Extravasation can cause
Acute hypophosphatemia: 0.15–0.33 mmol/ severe scaring.
kg/dose over 6 h until serum phosphorous is
greater than 2 mg/dL.
Propranolol

Maintenance/Replacement Neonates with thyroid storm:


Oral: 0.5–2 mg/kg/24 h divided into q6 to
IV 0.5–1.5 mmol/kg/24 h. 8 h.
Oral 2–3 mmol/kg/24 h. Children:
Recommended infusion rate: 0.1 mM/kg/h of Oral: Start at 0.5–1 mg/kg/24 h divided into
phosphate. When potassium salt is used, the rate q6 to 8 h; increase dosage q3 to 5 days prn.
will be limited by the maximum potassium Usual dosage range: 2–4 mg/kg/24 h divided
infusion rate. Do not co-infuse with calcium- into q6 to 8 h. Maximum dose: 10–40 mg/24 h or
containing products. 16 mg/kg/24 h.
Atenolol is suggested to be preferable to pro-
pranolol due to its more selective cardiac effects
Potassium Iodide but there is little experience in children. It is
reported to be used in 1–2 mg/kg per day as a
Neonatal Graves disease: 1–2 drops of Lugol’s single daily dose or given every 12 hours; maxi-
sol p.o., q8h. mum is reported to be 100 mg/dose.
Suggested Reading 383

Hypertension The doses arise from the differing recommen-


dations from various manufacturers for the indi-
Children: cations noted.
p.o.: Initial: 0.5–1 mg/kg/24 h divided into q6
to 12 h. May increase dose by 1 mg/kg q3 to 7
days prn. Noonan’s Syndrome
Maximum dose: 6 mg/kg/24 h divided into q6
to 12 h. Up to 0.066 mg/kg/day SC.
Adults:
p.o.: 40 mg/dose p.o., b.i.d., or 60–80 mg/
dose (sustained-release capsule) p.o., qd. May Prader–Willi Syndrome
increase by 10–20 mg/dose, q3 to 5 days.
Maximum dose reported: 640 mg/24 h 0.24 mg/kg/week SC.
although lower doses must be tried first.

Propylthiouracil: Black box warning: Not to be SGA


used in children due to risk of liver damage.
Up to 0.48 mg/kg/week or 0.033 mg/kg/day.

Sodium Bicarbonate SHOX Deficiency


To Correct Metabolic Acidosis BUT NOT 0.33–0.35 mg/kg/week SC.
RECOMMENDED FOR USE IN DKA OTHER
THAN EXCEPTIONAL CIRCUMSTANCES
Calculate patient’s dose with the following Turner Syndrome
formulas:
Neonates, infants, and children: 0.375 mg/kg/week SC dose depending upon the
HCO3– (mEq) = 0.3 × weight (kg) × base deficit manufacturer’s recommendations.
(mEq/L)
Adults:
HCO3− (mEq) = 0.2 × weight (kg) × base deficit Spironolactone
(mEq/L)
HCO3− (mEq) = 0.5 × weight (kg) × [24 − serum Diuretic
HCO3− (mEq/L)]
For either: Neonates: 1–3 mg/kg/24 h divided into qd to
HCO3− (mEq) = 0.5 × weight (kg) × [24 − serum b.i.d., p.o.
HCO3− (mEq/L)] Children: 1–3.3 mg/kg/24 h divided into qd to
q.i.d., p.o.
Adults: 25–200 mg/24 h divided into qd to
Somatropin (Growth Hormone) q.i.d., p.o.
Maximum dose: 200 mg/24 h.
Given 6–7 days per week (longer term prepara-
tions are soon to be available).
Primary Aldosteronism

Growth Hormone Deficiency Children: 125–375 mg/m2/24 h divided into b.i.d.


to q.i.d., p.o.
0.16–0.34 mg/kg/week SC dose depending upon Adults: 100–400 mg qd and titrate down for
the manufacturer’s recommendations. maintenance dosage.
384 15 Medications for Pediatric Endocrinology

Hirsutism or Acne in Women Subcutaneous/Intramuscular

Adults: 50–200 mg/24 h qd or divided into b.i.d., p.o. Children: 2.5–10 U, b.i.d. to q.i.d. as needed to a
Results may not appear for 6–12 months. maximum of 60 U per day.
Adults: 5–10 U, b.i.d. to q.i.d.
Continuous infusion (adults and children):
Testosterone Start at 0.5 milliunit/kg/h (0.0005 U/kg/h).
Double dosage every 30 min prn up to maximum
See Chap. 9. dose of 10 mU/kg/h (0.01 U/kg/h).

Vasopressin Suggested Reading

Diabetes Insipidus Dosages as presented in:

Branden Engorn, Jamie Flerlage, editors. The Harriet


DDAVP is the preferred treatment of diabetes
Lane handbook: a manual for pediatric house officers,
insipidus but in case the oral or nasal route is not the Harriet Lane Service, Children’s Medical and
appropriate the following is used. Titrate dose to Surgical Center of the Johns Hopkins Hospital, 20th
antidiuretic effect and watch for hypertension or ed. Elsevier; 2015.
Dosages from Thomson Truven Health MICROMEDEX.
hyponatremia.
Dosages from various textbook chapters or articles noted
in the other chapters.
Dosages from manufacturers’ information.
Laboratory Values for Pediatric
Endocrinology 16

This list includes some of the reference ranges laboratory values used most frequently in pediatric
endocrinology with sample preparation and sample sizes from Quest Diagnostics/Nichols Institute
and Esoterix/Labcorp. Other laboratories perform these tests and a few also have pediatric standards
(e.g., ARUP). However, we do not recommend using a laboratory that has not established pediatric
standards because the lower detectable limits, or the accurate area of the standard curves of the labora-
tory, might be well above pediatric values. Laboratories that are set up for pediatric endocrine samples
will accept smaller volumes than requested of adult patients. The minimum volumes listed here can
be used for smaller children but there may not be a duplicate test available to check results when such
small quantities are utilized; consult with the laboratory for details of the most recent collection size,
storage, and transport. You must check your results against the standards of the laboratory you choose,
as the laboratories recommend, as the reference ranges have changed during the development of this
text and will likely have been modified further since the time of publication of this book; the following
is only a guide.
Laboratories may combine tests into a panel for more efficient use of the sample, e.g., CAH
panel or hirsutism panel, so check with the laboratories for what panels are available. Panels are
offered from the major laboratories and provide the ability to collect small sample volumes for
multiple tests; if each individual test was ordered separately the volume might be prohibitive for
small children.

Adrenocorticotropic Hormone (ACTH) Quest

1.5 mL plasma in ethylenediaminetetraacetic acid (EDTA; lavender-top tube).


Minimum quantity: 0.5 mL.
Absolute minimum: 0.3 mL.
Transfer the plasma to a special transport tube, and ship frozen. Do not thaw.
Drawn at 7–10 a.m.

Adults 6–50 pg/mL


Dexamethasone suppressed 2–8 pg/mL
Children
3–17 years 9–57 pg/mL

© Springer International Publishing Switzerland 2016 385


D.M. Styne, Pediatric Endocrinology, DOI 10.1007/978-3-319-18371-8_16
386 16 Laboratory Values for Pediatric Endocrinology

Adrenal Antibody Screen with Reflex Titer QUEST

2 mL refrigerated serum.
0.5 mL serum
0.05 mL absolute minimum quantity
Adrenal antibody screen with reflex to titer
Red-top tube with no gel
0.5 mL minimum
0.05 mL absolute pediatric minimum
Negative
<1:10

Albumin (Microalbumin), 24-h Urine Quest

5 mL urine from 24-h urine collection; avoid glass container; 2 mL minimum in sterile screw-cap
container. Ship refrigerated.
Absolute minimum: 0.3 mL.
Record 24-h urine volume on test-request form and urine vial. Do not use preservatives. Ship
refrigerated.
Children and adults: ≤30 mg/24 h

Albumin Serum Quest

1 mL serum.
Minimum quantity: 0.5 mL.
Overnight fasting is preferred. Ship refrigerated.
3.6–5.1 g/dL.

Aldosterone, 24-h Urine Quest

5 mL urine from 24-h urine collection.


Minimum quantity: 0.8 mL.
Absolute minimum: 0.5 mL.
Collect urine with 10 g of boric acid to maintain pH <7.5. Record 24-h urine volume on test-request
form and urine vial. Refrigerate during collection and ship refrigerated.

Children
2–7 years <5.7 μg/24 h
8–11 years <10.2 μg/24 h
12–16 years <15.6 μg/24 h
Adults
2.3–21.0 μg/24 h

See creatinine standards as many urine test results are standardized to creatinine values.
Alkaline Phosphatase, Bone-Specific Labcorp 387

Aldosterone Serum, LC/MS/MS Quest

1 mL serum in no additive (red-top tube).


Minimum quantity: 0.25 mL.
Absolute minimum: 0.1 mL.
Draw “upright” samples ≥30 min after patient sits up. Ship refrigerated.

Children (random sodium diet)


Premature infants (31- to 35-week gestation) ≤144 ng/dL
Term infants, 3 days old ≤217 ng/dL
1–12 months 2–70 ng/dL
1–4 years 2–37 ng/dL
5–9 years <9 ng/dL
10–13 years <21 ng/dL
14–17 years <35 ng/dL
Tanner II–III
Males 1–13 ng/dL
Females 2–20 ng/dL
Tanner IV–V
Males 3–14 ng/dL
Females 4–32 ng/dL

Alkaline Phosphatase, Serum Quest

1 mL serum in no additive (red-top tube).


Minimum quantity: 0.5 mL.
Hemolyzed specimens are not acceptable. Ship frozen.

Male (U/L) Female (U/L)


<1 month 75–316 48–406
1–11 months 82–383 124–341
1–3 years 104–345 108–317
4–6 years 93–309 96–297
7–9 years 47–324 184–415
10–12 years 91–476 104–471
13–15 years 92–468 41–244
16–19 years 48–230 47–176

Alkaline Phosphatase, Bone-Specific Labcorp

2 mL frozen serum in no additive (red-top tube) allow serum to clot completely before centrifuging.
Minimum quantity: 1.0 mL.
Ship serum frozen in plastic screw top tube.
388 16 Laboratory Values for Pediatric Endocrinology

Age Male (μg/L) Female (μg/L)


0–5 months Not established Not established
6–11 months 48.2−170.2 50.4−154.0
1 year 41.9−229.6 44.2−178.3
2 years 41.7−139.4 34.9−195.4
3–4 years 37.2−127.7 37.4−96.8
5–6 years 42.0−104.2 31.4−100.8
7–8 years 59.0−101.3 44.0−135.8
9–10 years 50.4−133.0 47.9−150.8
11–12 years 53.3−156.8 24.2−133.3
13–14 years 77.5−169.8 19.8−92.7
15–16 years 26.8−173.4 11.2−30.2
17–18 years 15.4−69.8 8.8−29.0

Androstenedione LC/MS/MS Quest

1 mL room temperature serum in no additive (red-top tube).


Minimum quantity: 0.3 mL.
Absolute minimum: 0.15 mL
An early morning specimen is preferred. Ship refrigerated.

Children
Premature infants (31- to 35-week gestation) ≤480 ng/dL
Term infants, 3 days old ≤290 ng/dL
1–12 months 6–78 ng/dL
1–4 years 5–51 ng/dL
5–9 years 6–115 ng/dL
10–13 years 12–221 ng/mL
14–17 years 22–225 ng/mL
Tanner II–III
Males 17–82 ng/dL
Females 43–180 ng/dL
Tanner IV–V
Males 57–150 ng/dL
Females 73–220 ng/dL

Angiotensin II Quest

1 mL plasma in EDTA (lavender-top tube).


Minimum quantity: 0.3 mL.
Absolute minimum: 0.15 mL.
Freeze within 1 h of collection. Ship frozen.
Adults: <86 ng/L.
Children: No data available.
C Peptide, Serum Quest 389

Anti-Mullerian Hormone (AMH) Quest

1 mL frozen serum
Absolute minimum: 0.5 mL

Male
<1 year 37.20–345.67 ng/mL
1–6 years 59.54–320.65 ng/mL
7–11 years 40.99–203.67 ng/mL
12–17 years <128.29 ng/mL
≥18 years 1.15–15.23 ng/mL
Female
<14 years 0.49–3.15 ng/mL
14–19 years 1.28–16.37 ng/mL
20–29 years 0.76–11.34 ng/mL
30–39 years <9.24 ng/mL
40–49 years <4.50 ng/mL
≥50 years <0.45 ng/mL

Arginine Vasopressin Quest

4 mL plasma in EDTA (lavender-top tube).


Minimum quantity: 1.1 mL.
Absolute minimum: 0.55 mL
Draw blood in prechilled lavender-top tube. Transport in an ice bath to a refrigerated centrifuge.
Separate and freeze immediately. Do not thaw. Ship frozen.
Children and adults: 1.0–13.3 pg/mL (2.5 pg = 1 μU).

Congenital Adrenal Hyperplasia 21-Hydroxylase Deficiency Common


and Rare Mutations Quest

Collected lavender-top tube 3 mL minimum.

C Peptide, Serum Quest

2 mL frozen serum.
Minimum quantity: 0.5 mL.
Overnight fasting is required. Ship frozen.
Adults: 0.8–3.1 ng/mL. Values will vary based upon blood sugar.
390 16 Laboratory Values for Pediatric Endocrinology

Calcitonin Quest

1 mL frozen serum in no additive (red-top tube).


Minimum quantity: 0.5 mL.
Absolute minimum: 0.5 mL
Overnight fasting is preferred. Ship frozen.

<6 months <41 pg/mL


6 months to 3 years <14 pg/mL
3–17 years <6 g/mL

Calcium, 24-h Urine Quest

10 mL urine from a 24-h urine collection.


Minimum quantity: 2 mL.
Collect urine with 25 mL of 6 N HCl to maintain a pH <3. Record 24-h urine volume on test-request
form and urine vial. Ship refrigerated.

Adults
Males 50–300 ng/24 h
Females 50–250 ng/24 h
Children mg/mg creatinine
1–11 months 0.03–0.81
1 year 0.032–0.56
2 years 0.0–0.50
3–4 years 0.022–0.41
5–6 years 0.01–0.30
7–9 years 0.01–0.25
10–17 years 0.01–0.24

Calcium Ionized, Serum Quest

2 mL serum in no additive (red-top tube).


Minimum 0.6 mL
Absolute minimum: 0.3 mL.
Collect in gel-barrier tube, let clot, and spin immediately with cap on. Ship the unopened gel-barrier
tube refrigerated, so sample is not exposed to dry ice. Do not open. Ship refrigerated.

Children
8 months to 10 years 4.9–5.4 mg/dL
11–17 years 4.8–5.3 mg/dL
Adults
4.8–5.6 mg/dL
Catecholamines, Fractionated, 24-h Urine Quest 391

Calcium, Serum Quest

1 mL serum in no additive (red-top tube).


Minimum quantity: 0.5 mL.
Absolute minimum: 0.2 mL.
Overnight fasting is preferred. Ship refrigerated.

Children
<1 month 8.4–10.6 mg/dL
1–11 months 8.7–10.5
1–3 years 8.5–10.6
4–19 years 8.9–10.4 mg/dL

Carnitine, Serum Quest

3 mL serum in no additive (red-top tube).


Minimum quantity: 1 mL.
Serum or plasma should be removed from cells immediately after collection. Ship frozen.

Children (M)
Carnitine total Carnitine free Carnitine esters
1–17 years females 28–59 19–48 3–16
1–17 years males 32–62 25–54 4–12

Carotene Quest

1 mL serum in no additive (red-top tube).


Minimum quantity: 0.5 mL.
Separate from cells as soon as possible after clotting. Wrap tube in aluminum foil to protect from
light. Overnight fasting is preferred. Ship refrigerated.

Children
9 months to 6 years 5–80 μg/dL
7–17 years 9–190 μg/dL

Catecholamines, Fractionated, 24-h Urine Quest

Includes dopamine, epinephrine, and norepinephrine.


10 mL urine from a 24-h urine collection.
Minimum quantity: 5 mL.
Collect urine with 25 mL of 6 N HCl to maintain a pH <3. Urine without preservatives is accept-
able if pH is <6 and the sample is shipped frozen. Record 24-h urine volume on test-request
392 16 Laboratory Values for Pediatric Endocrinology

form and urine vial. It is preferable for the patient to be off medication for 3 days before collection.
Ship refrigerated.

Epinephrine
3–8 years 1–7 μg/24 h
9–12 years ≤8 μg/24 h
13–17 years ≤11 μg/24 h
Adults 2–24 μg/24 h
Norepinephrine
3–8 years 5–41 μg/24 h
9–12 years 5–50 μg/24 h
13–17 years 12–88 μg/24 h
Adults 15–100 μg/24 h
Dopamine
3–8 years 80–378 μg/24 h
9–12 years 51–474 μg/24 h
13–17 years 51–645 μg/24 h
Adults 52–480 μg/24 h
Total (N + E)
3–8 years 9–51 μg/24 h
9–12 years 9–71 μg/24 h
13–17 years 13–90 μg/24 h
Adults 26–121 μg/24 h

Catecholamines, Fractionated, Plasma Quest

Includes dopamine, epinephrine, and norepinephrine.


4 mL plasma in sodium heparin (green-top tube).
Minimum quantity: 2.5 mL.
Absolute minimum: 1 mL.
Vacutainer is to be chilled before venipuncture. Draw sample as follows: Insert catheter in patient’s
vein. Instruct patient to lie down for 30 min with catheter in place, and then draw supine specimen
in green-top tube. Chill specimen immediately in ice water. Next, instruct patient to sit up for
15 min with catheter still in place. Draw upright specimen in green-top tube. Chill specimen imme-
diately in ice water. Plasma should be separated in a refrigerated centrifuge within 30 min of col-
lection and then frozen immediately at −20 °C in plastic vials. Plasma must stay frozen. Thawed
samples are unacceptable. Overnight fasting is required. Ship frozen.

Adults

Supine Upright
Dopamine <10 pg/mL <20 pg/mL
Epinephrine <50 pg/mL <95 pg/mL
Norepinephrine 112–658 pg/mL 217–1,109 pg/mL
Total (N + E) 123–671 pg/mL 242–1,125 pg/mL
Catecholamines, Fractionated, Random Urine Quest 393

Children supine
Plasma catecholamine measurements are generally unreliable because of stress in infants and small
children. Urinary catecholamine assays are more reliable.

Age 3–15 years


Dopamine <60 pg/mL
Epinephrine ≤464 pg/mL
Norepinephrine ≤1,251 pg/mL

Catecholamines, Fractionated, Random Urine Quest

Includes epinephrine, norepinephrine, and dopamine.


10 mL urine in sterile screw-cap container.
Minimum quantity: 5 mL.
After urine collection, add 6 N HCl to maintain a pH <3. Urine without preservative is acceptable if
pH is <6 and the sample is shipped frozen. It is preferable for the patient to be off alcohol, coffee,
tea, tobacco and strenuous exercise as well as certain medications if possible for 3 days before
collection.

Epinephrine
Birth to 6 months 2–45 μg/g Cr
7–11 months 5–45 μg/g Cr
1–2 years 1–49 μg/g Cr
3–8 years 4–32 μg/g Cr
9–12 years 1–15 μg/g Cr
13–17 years 1–10 μg/g Cr
Adults 2–16 μg/g Cr
Norepinephrine
Birth to 6 months 12–286 μg/g Cr
7–11 months 19–250 μg/g Cr
1–2 years 25–210 μg/g Cr
3–8 years 20–108 μg/g Cr
9–12 years 20–73 μg/g Cr
13–17 years 15–58 μg/g Cr
Adults 7–65 μg/g Cr
Total N + NE
Birth to 6 months 24–322 μg/g Cr
7–11 months 10–295 μg/g Cr
1–2 years 30–263 μg/g Cr
3–8 years 30–130 μg/g Cr
9–12 years 25–90 μg/g Cr
13–17 years 20–70 μg/g Cr
Adults 9–74 μg/g Cr
(continued)
394 16 Laboratory Values for Pediatric Endocrinology

(continued)
Dopamine
Birth to 6 months 107–2,180 μg/g Cr
7–11 months 96–2,441 μg/g Cr
1–2 years 86–1,861 μg/g Cr
3–8 years 295–1,123 μg/g Cr
9–12 years 164–744 μg/g Cr
13–17 years 156–551 μg/g Cr
Adults 40–390 μg/g Cr

Corticosterone LC/MS/MS Quest

1 mL serum in no gel or additive (red-top tube); early morning specimen is best.


Minimum quantity: 0.25 mL.
Absolute minimum: 0.10.
Ship refrigerated.
Adults
8–10:00 a.m.: 59–1,293 ng/dL.
4–6:00 p.m.: ≤386 ng/dL.

1–12 months
Males 78–1,750 ng/dL
Females 89–1,200 ng/dL
1–4 years
Males 120–1,290 ng/dL
Females 160–2,040 ng/dL
5–9 years
Males <677 ng/dL
Females <677 ng/dL
10–13 years
Males <1,420
Females <1,420
14–17 years
Males <1,238
Females <1,238
Tanner II–III
Males 115–1,220 ng/dL
Females 110–600 ng/dL
Tanner IV–V
Males 165–840 ng/dL
Females 160–390 ng/dL
Cortisol-Free Serum Quest 395

Corticotropin-Releasing Hormone (Mainly Use for Ectopic Production


of CRH) Quest

3 mL plasma into a sodium heparin green top tube.


Minimum quantity: 1.1 mL.
Do not thaw. Ship frozen.

Men and women ≤42 pg/mL


Pregnancy
First trimester ≤40 pg/mL
Second trimester ≤153 pg/mL
Third trimester ≤847 pg/mL
Cord blood ≤338

Cortisol-Binding Globulin (Transcortin) Quest

1 mL serum in no additive (red-top tube).


Minimum quantity: 0.2 mL.
Ship refrigerated.

Preterm infants younger than 8 days 6–26 mg/L


Adults 19–45 mg/L

Cortisol Urinary Free 24-h Quest

10 mL urine from a 24-h urine collection.


Minimum quantity: 2.6 mL.
Collect urine with 10 g of boric acid or keep urine refrigerated during collection if preservative is not
used. Record 24-h urine volume on test-request form and urine vial.

Adults
Males and females 4–50 μg/24 h
Children
1–4 years 0.9–8.2 μg/24 h
5–9 years 1.0–30.0 μg/24 h
10–13 years 1.0–45 μg/24 h
14–17 years 3.0–55 μg/24 h

Cortisol-Free Serum Quest

2 mL serum in no additive (red-top tube).


Minimum quantity: 0.7 mL.
396 16 Laboratory Values for Pediatric Endocrinology

Absolute minimum: 0.4 mL.


Grossly hemolyzed specimens are unacceptable. Ship refrigerated.

Adults
8–10:00 a.m. 0.07–0.93 μg/dL
4–6:00 p.m. 0.04–0.45 μg/dL
10.00–11:00 p.m. 0.04–0.35 μg/dL

Cortisol, Salivary

0.5 mL refrigerated saliva


Minimum quantity: 0.2 mL
Absolute minimum: 0.1 mL
Collected in a salivette tube 10 min after rinsing mouth with water. No oral intake (or smoking) for
1 h before collection. Blood from brushing teeth can affect result.

Adults
8–10 a.m. 0.04–0.56
4–6 p.m. <0.15
10–11 p.m. <0.09

Cortisol Serum Total LC/MS/MS Esoterix

0.5 mL serum plasma: Serum: Tigertop (SST) tube or redtop tube or Plasma: Lavendertop (EDTA)
tube.
Minimum quantity: 0.1 mL.
Specify the time of day specimen was collected. Grossly hemolyzed specimens are unacceptable.
Ship refrigerated.

Age Range (μg/dL)


Premature (26–28 weeks) Day 4 1.0–11
Premature (31–35 weeks) Day 4 2.5–9.1
Full-term Day 3 1.7–14
Full-term Day 7 2.0–11
31 days to 11 months 2.8–23
12 months to 15 years (8:00 a.m.) 3.0–21
8:00 a.m. 4:00 p.m.
Adults 8.0–19 4.0–11

Response to ACTH stimulation


Cortisol baseline 5–21 μg/dL
Cortisol 30 min 14–36 μg/dL
Cortisol 60 min 14–41 μg/dL
Creatinine Clearance Quest 397

Creatinine 24-h Urine Quest

10 mL with urine in 24-h urine container collection.


Minimum quantity: 0.5 mL.
Record 24-h urine volume on test-request form and urine vial. Ship refrigerated. No preservative.

Adults
Males and females 0.63–2.5 g/24 h
Children
3–8 years 0.11–0.68 g/24 h
9–12 years 0.17–1.41 g/24 h
13–17 years 0.29–1.87 g/24 h
>17 years 0.63–2.50 g/24 h

Creatinine Random Urine Quest

10 mL urine in sterile screw-cap container.


Minimum quantity: 2 mL.
Ship refrigerated.

Adults
Males and females 0.27–3.00 g/L
Children
≤6 months 2–32 mg/dL
7–11 months 2–36 mg/dL
1–2 years 2–128 mg/dL
3–8 years 2–149 mg/dL
9–12 years 2–183 mg/dL
>12 years male 20–370 mg/dL
>12 years female 20–320 mg/dL

Creatinine Clearance Quest

Includes serum and urine creatinine.


10 mL urine from a 24-h urine collection and 1 mL serum in no additive (red-top tube).
Minimum quantity: 0.5 mL serum and urine.
Record patient height, weight, and total 24-h urine volume on test-request form and urine vial. Serum
must be collected within 24 h of urine collection. Ship refrigerated.

Adults
Males 85–125 mL/min/1.73 m2 surface area (SA)
Females 75–115 mL/min/1.73 m2 SA
398 16 Laboratory Values for Pediatric Endocrinology

Creatinine Serum Quest

2 mL serum in no additive (red-top tube).


Minimum quantity: 0.2 mL.
Hemolyzed specimens are not acceptable. Ship refrigerated.

Adults
Male Female
0.6–1.4 mg/dL 0.6–1.4 mg/dL
Children
Age Male (mg/dL) Female (mg/dL)
≤2 days 0.79–1.58 0.79–1.58
3–27 days 0.35–1.23 0.35–1.23
1 month to 9 years 0.20–0.73 0.20–0.73
10–12 years 0.30–0.78 0.30–0.78
13–15 years 0.40–1.05 0.40–1.00
16–17 years 0.60–1.20 0.50–1.00
18–19 years 0.60–1.26 0.50–1.00

DHEA Serum Quest Esoterix

0.5 mL serum in no additive SST or (red-top tube).


Minimum quantity: 0.3 mL.
Specify age and sex on test-request form. Overnight fasting is preferred. Ship refrigerated.

Children
Premature infants
26–31 weeks 82–1,484 ng/dL
32–35 weeks 56–1,853 ng/dL
Full-term infants
2–7 days 41–1,292 ng/dL
8 days to 5 months <948 ng/dL
6–12 months <136 ng/dL
Prepubertal children
1–5 years <68 ng/dL
6–7 years <111 ng/dL
8–10 years <186 ng/dL
Pubertal children and adults
11–12 years <202 ng/dL
13–14 years <319 ng/dL
15–16 years 39–481 ng/dL
17–19 years 40–491 ng/dL
Deoxycorticosterone (DOC), Serum Esoterix 399

DHEA Sulfate, Serum Esoterix

0.5 mL serum in no additive (red-top tube).


Minimum quantity: 0.1 mL.
Specify age and sex on test-request form. Ship refrigerated.

Range (μg/dL)
Premature infants
26–28 weeks, day 4 123–882
31–35 weeks, day 4 122–710
Full-term infants
3 days 88–356
1–11 months DHEA-S levels fall to <112 μg/dL during the first month and decrease further
to <49 μg/dL by 6 months of age
Prepubertal children
1–5 years <57
6–7 years <72
8–10 years <193

Puberty
Tanner stage Age (years) Range (μg/dL) Tanner stage Age (years) Range (μg/dL)
Male Female
1 <9.8 13–83 1 <9.2 19–144
2 9.8–14.5 42–109 2 9.2–13.7 34–129
3 10.7–15.4 48–200 3 10.0–14.4 32–226
4 11.8–16.2 102–385 4 10.7–15.6 58–260
5 12.8–17.3 120–370 5 11.8–18.6 44–248

Deoxycorticosterone (DOC), Serum Esoterix

Age Range (ng/dL)


Premature (26–28 weeks) Day 4 20–105
Premature (34–36 weeks) Day 4 28–78
Newborn Levels are markedly elevated at birth and decrease
rapidly during the first week to the range of 7–49
as found in older infants.
1–11 months 7–49
Prepubertal children 2–34
Pubertal children and adults 8:00 a.m. 2–19
400 16 Laboratory Values for Pediatric Endocrinology

11-Deoxycortisol (Compound S) Corticosterone, Serum Esoterix

Eso range (ng/dL)


Premature infants
26–28 weeks, day 4 110–1,376
31–35 weeks, day 4 48–579
Full-term infants
3 days 13–147
31 days to 11 months <10–156
Prepubertal children
8:00 a.m. 20–155
Pubertal children and adults
8:00 a.m. 12–158

Dihydrotestosterone Esoterix

0.3 mL serum in no additive (red-top tube).


Minimum quantity: 0.1 mL.
Specify age and sex on test-request form. Ship refrigerated.

Male range (ng/dL) Female range (ng/dL)


Premature infants 10–53 2–13
Full-term newborns 5–60 <2–15
2 weeks to 6 months DHT decreases rapidly in the first week, and Levels decrease during the first
then increases to 12–85 ng/dL between 30 and month to < 3 ng/dL and remain
60 days. Levels then decrease gradually to there until puberty.
prepubertal values by 7 months.
Prepubertal children 1–10 years: <3 1–9 years: <3

Puberty
Tanner stage Age (years) Range (ng/dL) Tanner stage Age (years) Range (ng/dL)
Male Female
1 <9.8 <3 1 <9.2 3
2 9.8–14.5 3–17 2 9.2–13.7 5–12
3 10.7–15.4 8–33 3 10.0–14.4 7–19
4 11.8–16.2 22–52 4 10.7–15.6 4–13
5 12.8–17.3 24–65 5 11.8–18.6 3–18

Range (ng/dL)
Adults
Male 30–85
Female 4–22
Estradiol, Serum Esoterix 401

Dihydrotestosterone, Free, Serum Quest

Includes total, free, and percentage free dihydrotestosterone (DHT).


5 mL serum in no additive (red-top tube).
Minimum quantity: 2.5 mL.
Ship refrigerated.
% Free
Postpubertal males: 0.62–1.10 %.
DHT, free
Postpubertal males: 1.00–6.20 pg/mL.
Postpubertal females: 0.30–1.90 pg/mL.

Estradiol, Serum Esoterix

3 mL serum in no additive (red-top tube).


Minimum quantity: 1.2 mL.
Separate serum within 1 h; transfer into a plastic transport tube.
Specify age and sex on test-request form. Ship refrigerated frozen.

Range (pg/mL)
Newborn Levels are markedly elevated at birth and fall rapidly during the first week to
prepubertal values of <15 pg/mL.
1–6 months
Male Levels increase to 10–32 pg/mL between 30 and 60 days, and then decline to
prepubertal levels of <15 pg/mL by 6 months.
1–11 months
Female Levels increase to 5.0–50 pg/mL between 30 and 60 days, and then decline to
prepubertal levels of <15 pg/mL during the first year.
Prepubertal children
Males (1–10 years) <15
Females (1–9 years) <15

Puberty
Tanner stage Age (years) Range (μg/dL) Tanner stage Age (years) Range (μg/dL)
Male Female
1 <9.8 5.0–11 1 <9.2 5.0–20
2 9.8–14.5 5.0–16 2 9.2–13.7 10–24
3 10.7–15.4 5.0–25 3 10.0–14.4 7.0–60
4 11.8–16.2 10–36 4 10.7–15.6 21–85
5 12.8–17.3 10–36 5 11.8–18.6 34–170

Adults Range (pg/mL)


Male 8–35
Female
Follicular 30–100
Luteal 70–300
Postmenopausal <15
402 16 Laboratory Values for Pediatric Endocrinology

Estrone, Serum Esoterix

3 mL serum in no additive (red-top tube).


Minimum quantity: 1.2 mL.

Puberty
Tanner stage Age (years) Range (μg/dL) Tanner stage Age (years) Range (μg/dL)
Male Female
1 <9.8 5.0–17 1 <9.2 4.0–29
2 9.8–14.5 10–25 2 9.2–13.7 10–33
3 10.7–15.4 15–25 3 10.0–14.4 15–43
4 11.8–16.2 15–45 4 10.7–15.6 16–77
5 12.8–17.3 20–45 5 11.8–18.6 29–105

Adults Range (pg/mL)


Male 10–50
Female
Follicular 30–100
Luteal 90–160
Postmenopausal <40

Follicle-Stimulating Hormone, Third Generation, Serum Esoterix

1 mL serum in no additive (red-top tube).


Minimum quantity: 0.2 mL, not for eso
Ship refrigerated.
Eso range (mIU/mL)

Infants
Male Female
4 weeks to 11 months 0.16–4.1 0.24–14.2
Levels are for infants from 4 weeks Levels are for infants from 4 weeks of
of age to 1 year. FSH in males declines age to 1 year. FSH declines more slowly
to prepubertal levels by the end of the than in males to reach prepubertal levels
first year. by the end of the second year.
Prepubertal children
Male Female
12 months to 8 years 0.26–3.0 1.0–4.2
Gastrin Serum Quest 403

Puberty
Tanner Age Range Mean Tanner Age Range Mean
stage (years) (pg/dL) (μg/dL) stage (years) (pg/dL) (μg/dL)
Male Female
1 <9.8 0.26–3.0 0.98 1 <9.2 1.0–4.2 2.1
2 9.8–14.5 1.8–3.2 2.5 2 9.2–13.7 1.0–10.8 4.0
3 10.7–15.4 1.2–5.8 2.9 3 10.0–14.4 1.5–12.8 5.1
4 11.8–16.2 2.0–9.2 4.4 4 10.7–15.6 1.5–11.7 6.4
5 12.8–17.3 2.6–11.0 6.1 5 11.8–18.6 1.0–9.2 4.9

Adults Range (pg/mL)


Males
20–50 years 2.0–9.2
Females
Follicular and luteal 1.8–11.2
Midcycle 6–35
Postmenopausal 30–120

Fructosamine, Serum Quest

1 mL serum in no additive (red-top tube).


Minimum quantity: 0.5 mL.
0.2 absolute minimum.
Ship refrigerated.

Adults
Males and females 190–270 μM

Gastrin Serum Quest

2 mL serum.
Minimum quantity: 0.5 mL.
Absolute minimum 0.175 mL
Overnight fasting is required. Ship frozen.

Adults
Males and females <100 pg/mL
Children
5–17 years 13–64 pg/mL
404 16 Laboratory Values for Pediatric Endocrinology

Glucagon Quest

3 mL plasma in EDTA (lavender-top tube).


Minimum quantity: 1.1 mL.
Absolute minimum 0.55 mL.
Do not thaw. Overnight fasting is required. Ship frozen.

Adults <134 pg/mL


Children
Cord blood <2:15
Day 1 <240
Day 2 <400
Day 3 <420
Day 4–14 <148

Glucose, Serum

1 mL serum in no additive (red-top tube), but grey top at local lab is best.
Minimum quantity: 0.2 mL.
Overnight fasting is preferred. Ship refrigerated.

Children and adults


Fasting 65–100 mg/dL
Impaired fasting glucose 101–125 mg/dL (ADA Guidelines)
Provisional diagnosis of diabetes ≥126 mg/dL fasting or >200 mg/dL casual, non-fasting
(ADA Guidelines)

Glutamic Acid Decarboxylase-65 Autoantibodies Quest

1 mL serum in no additive (red-top tube).


Minimum quantity: 0.2 mL.
Absolute minimum 0.04 mL.
Ship frozen.
Children and adults: Positive result, ≥1.0 U/mL.

Glycated Albumin Quest

3 mL plasma in EDTA (lavender-top tube).


Minimum quantity: 2 mL.
Absolute minimum 0.3 mL.
Spin and separate plasma immediately. Overnight fasting is required. Ship refrigerated.
Children and adults: 0.8–1.4 %.
Growth Hormone-Binding Protein (GHBP) Esoterix 405

1,5-Anhydroglucitol Esoterix

1 mL room-temperature serum in a red-top tube without gel or SST.


Minimum 0.75 mL.

Age Range (μg/mL)


<18 years Not established
Adult males 10.7–32.0
Adult females 6.8–29.3
Glycemic control goal for diabetic patients: >10

Growth Hormone, Serum Esoterix

1 mL serum in no additive (red-top tube).


Minimum quantity: 0.4 mL.
Ship refrigerated.

Newborn Eso range (ng/mL)


1 day 5–53
2–7 days 5–27
31 days to 11 months 2–10
Following an 8–12-h overnight fast:
Children <6
Adults <6

Adults: ≤10.0 ng/mL.


Children: ≤20 years ≤13 ng/mL.
Because of diurnal variability, random human growth hormone (hGH) values are not reliable for diag-
nosis of GH deficiency or acromegaly. Values are quite elevated for several days after birth. See
Chap. 5 for instructions on GH testing.

Growth Hormone Antibody Quest

0.5 mL serum in no additive (red-top tube).


Minimum quantity: 0.2 mL.
Absolute minimum 0.03 mL.
Ship refrigerated.
Children and adults: Negative.

Growth Hormone-Binding Protein (GHBP) Esoterix

1 mL serum in no additive (red-top tube).


Minimum quantity: 0.2 mL.
Ship frozen.
406 16 Laboratory Values for Pediatric Endocrinology

Range (pmol/L)
Children
<1 year <125–762
2–9 years 267–1,638
10–14 years 431–1,892
Adults
20–50 years 686–2,019
Laron dwarfism <300

Growth Hormone-Releasing Hormone Quest

4 mL plasma in parathyroid hormone-related protein and releasing factor tube.


Minimum quantity: 1.1 mL.
Use special collection tube labeled parathyroid hormone RP and releasing factor tube, available from
Quest Diagnostics Incorporated, Nichols Institute. Do not thaw. Ship frozen.

Adults
Males and females ≤49 pg/mL
Children
4–14 years ≤19 pg/mL

Hemoglobin A1C, Blood Quest

1 mL whole blood in EDTA (lavender-top tube).


Minimum quantity: 0.5 mL.
Absolute minimum 0.15 mL.
Transfer whole blood to plastic shipping vial to prevent breakage. Avoid freezing and thawing. Ship
refrigerated.

Children and adults although children have more


variable results
Nondiabetic <5.7 %
Increased diabetes risk 5.7–6.0 %
High diabetes risk 6.1–6.4 %
Diabetes ≥6.5
Reevaluate therapy >8 %

Homovanillic Acid, 24-h Urine Quest

10 mL urine from 24-h urine collection.


Minimum quantity: 5 mL.
Absolute minimum 1.1 mL.
18-Hydroxycorticosterone, Serum Esoterix 407

Collect urine with 25 mL of 6 N HCl to maintain a pH <3. Urine without preservative is acceptable
if pH is <6 and the sample is shipped frozen. Record 24-h urine volume on test-request form and urine
vial. It is preferable for the patient to be off medications for 3 days before collection. Ship
refrigerated.

Adults
Males and females 1.6–7.5 mg/24 h
Children
3–8 years 0.5–6.7 mg/24 h
9–12 years 1.1–6.8 mg/24 h
13–17 years 1.4–7.2 mg/24 h

17-Hydroxycorticosteroids, 24-h Urine Quest

20 mL urine from 24-h urine collection.


Minimum quantity: 5 mL.
Collect urine with 10 g of boric acid to maintain pH <7.5. Record 24-h urine volume on test-request
form and urine vial. Ship refrigerated.

Adults
Males 4–11 mg/24 h
Females 3–10 mg/24 h
Children
1–2 years 0.5–2.5 mg/24 h
3–4 years 1.0–4.0 mg/24 h
5–6 years 1.0–4.8 mg/24 h
7–8 years 1.0–5.6 mg/24 h
9–10 years 1.0–7.0 mg/24 h
11–12 years 1.5–8.0 mg/24 h
Males
13–16 years 2.0–6.0 mg/24 h
17–20 years 3.0–10.0 mg/24 h
Females
13–16 years 2.8–6.8 mg/24 h
17–20 years 2.0–7.0 mg/24 h

18-Hydroxycorticosterone, Serum Esoterix

3 mL serum in no additive (red-top).


Minimum quantity: 1.0 mL.
408 16 Laboratory Values for Pediatric Endocrinology

18-OH-Corticosterone
Range (ng/dL)
Premature infants
26–28 weeks, day 4 10–670
31–35 weeks, day 4 57–410
Full-term infants
3 days 31–546
31 days to 11 months 5–220
Children
12–23 months 18–155
24 months to 9 years 6–85
10 to 14 years 10–72
Adults 9–58

8:00 a.m. Supine: 4–21.


8:00 a.m. Upright: 5–46.

5-Hydroxyindoleacetic Acid (5-HIAA), 24-h Urine Quest

10 mL urine from 24-h urine collection.


Minimum quantity: 5 mL.
Collect urine with 25 mL of 6 N HCl to maintain a pH <3. Urine without preservative is acceptable if
pH is <6 and the sample is shipped frozen. Keep urine refrigerated during collection if preservative
is not used. Record 24-h volume on test-request form and urine vial.
2–10 years: ≤8.0 mg/24 h: or ≤12 mg per gram creatinine.
<10 years: ≤6.0 mg/24 h: or ≤10 mg per gram creatinine.

5-Hydroxyindoleacetic Acid (5-HIAA), Random Urine Quest

10 mL urine in sterile screw-cap container.


Minimum quantity: 5 mL.
After urine collection, add 0.5–1.0 g boric acid (or 6 N HCl) to maintain a pH <3. Urine without pre-
servative is acceptable if pH is <6 and the sample is shipped frozen. Keep urine refrigerated during
collection if preservative is not used. Record patient’s age on test-request form and urine vial. Ship
refrigerated.
2–10 years: ≤12 mg/g Cr.
>10 years: ≤10 mg/g Cr.

17-Hydroxypregnenolone, Serum Esoterix

1 mL serum in no additive (SST).


Minimum quantity: 0.1 mL.
17-Hydroxyprogesterone (17OHP), Serum Esoterix 409

Range (ng/dL)
Premature infants
26–28 weeks, day 4 375–3,559
31–35 weeks, day 4 64–2,380
Full-term infants
3 days 10–829
1–5 months 36–763
6–11 months 42–540
Prepubertal children
12–23 months 14–207
24 months to 5 years 10–103
6–9 years 10–186
Pubertal age groups 44–235
Adults 53–357

17-Hydroxyprogesterone (17OHP), Serum Esoterix

1 mL serum in no additive (red-top or SST).


Minimum quantity: 0.1 mL.
Ship in plastic transport tube; ship frozen.

Range (ng/dL)
Premature infants
26–28 weeks, day 4 124–841
31–35 weeks, day 4 26–568
Full-term infants
Day 3 <78
Male 1–11 months Levels increase after the first week to peak values ranging from
40 to 200 ng/dL between 30 and 60 days. Levels then decline to
a prepubertal value of <91 before 1 year.
Female 1–11 months 13–106
Prepubertal children
Males (1–10 years) <91
Females (1–9 years) <91

Puberty
Tanner Age Range Mean Tanner Age (years) Range Mean
stage (years) (μg/dL) (μg/dL) stage (μg/dL) (μg/dL)
Male Female
1 <9.8 <91 38 1 <9.2 <83 31
2 9.8–14.5 <116 51 2 9.2–13.7 11–98 49
3 10.7–15.4 10–138 57 3 10.0–14.4 11–155 70
4 11.8–16.2 29–180 80 4 10.7–15.6 18–230 91
5 12.8–17.3 24–175 97 5 11.8–18.6 20–265 108
410 16 Laboratory Values for Pediatric Endocrinology

Adults Range (pg/mL)


Male 27–199
Female
Follicular 15–70
Luteal 35–290

Insulin-Like Growth Factor 1 (IGF-1) (Previously Called Somatomedin C)


Esoterix

1 mL serum in no additive (red-top tube or SST).


Minimum quantity: 0.5 mL.
Ship refrigerated.

Age Term Preterm


Range (ng/mL) Mean (ng/mL) Range (ng/mL) Mean (ng/mL)
Birth 15–109 59 21–93 51
2 months 15–109 55 23–163 81
4 months 7–124 50 23–171 74
6 months 7–93 41 15–132 61
12 months 15–101 56 15–179 77
Male Female
Age Range (ng/mL) Mean (ng/mL) Range (ng/mL) Mean (ng/mL)
1–2 years 30–122 76 56–144 100

Female Tanner 1 Tanner 2 Tanner 3 Tanners 4 and 5


Age Range Mean Range Mean Range Mean Range Mean
(years) (ng/mL) (ng/mL) (ng/mL) (ng/mL) (ng/mL) (ng/mL) (ng/mL) (ng/mL)
3 26–162 77
4 32–179 88
5 39–198 100
6 47–217 113
7 55–238 127
8 64–259 142 89–369 181
9 74–282 158 96–399 196 192–568 350
10 85–306 175 104–431 212 192–568 350 279–664 446
11 97–332 194 112–466 229 192–568 350 268–646 433
12 110–358 213 121–504 247 192–568 350 248–612 406
13 131–545 267 192–568 350 229–579 380
14 136–566 278 192–568 350 211–547 356
15 192–568 350 194–516 332
16 177–487 309
17 162–458 287
18 147–430 266
Total N Tanner 1: 138 Tanner 2: 158 Tanner 3: 114 Tanners 4 and 5: 155
subjects
IGF-Binding Protein-2 (IGFBP-2) Quest 411

IGF-2 Esoterix

0.5 mL plasma in EDTA (SST).


Minimum quantity: 0.1 mL.
Overnight fasting is preferred. Ship frozen in plastic transfer tube.

Range (ng/mL) Mean


Prepubertal 334–642 488
Pubertal 245–737 491
Adults 288––736 512

IGF-Binding Protein-1 (IGFBP-1) Quest

1 mL serum in no additive (red-top tube).


Minimum quantity: 0.2 mL.
Absolute minimum 0.025 mL.
Overnight fasting is preferred. Ship frozen.

Adults
Males and females 5–34 ng/mL
Children
5–9 years 15–95 ng/mL
10–14 years 8–64 ng/mL
15–18 years 5–40 ng/mL

IGF-Binding Protein-2 (IGFBP-2) Quest

1 mL serum in no additive (red-top tube).


Minimum quantity: 0.2 mL.
Absolute minimum 0.25 mL.
Ship frozen.

Adults
18–49 years 38–267 ng/mL
>49 years 47–350 ng/mL
Children
5–9 years 49–208 ng/mL
10–13 years 41–167 ng/mL
14–17 years 37–135 ng/mL
412 16 Laboratory Values for Pediatric Endocrinology

IGF-Binding Protein-3 (IGFBP-3) Esoterix

1 mL serum in no additive (red-top tube or EDTA lavender top).


Minimum quantity: 0.1 mL.
Ship frozen.

Range (mg/L) Mean (mg/L)


Premature infants
<1 month 0.3–1.4 0.9
2–3 months 0.9–2.3 1.6
4–5 months 0.4–2.2 1.5
6–11 months 1.0–2.3 1.5
Full-term infants
<1 month 0.4–1.7 0.9
2–3 months 0.5–2.1 1.3
4–5 months 0.6–2.4 1.4
6–11 months 0.5–2.4 1.4
Children
12 months to 4 years 0.8–3.0 2.1
5–6 years 1.5–3.4 2.4
7–8 years 2.1–4.2 3
9–11 years 2.0–4.8 3.3
12–13 years 2.1–6.2 3.8
14–15 years 2.2–5.9 4.2
16–18 years 2.5–4.8 3.8

Inhibin B Quest

1 mL frozen serum and red-top tube with no gel.


0.5 mL absolute minimum.

Men 47–308 pg/mL


Women
Premenopausal <153 pg/mL
Postmenopausal <10 pg/mL

Children
Males Females
5–9.9 years 21–166 <18
10–13.9 years 41–328 <86
14–17.9 years 135–368 <123
Leptin, Serum Quest 413

Insulin Antibodies, Highly Sensitive Quest

For detection of insulin autoantibody in patients not receiving insulin therapy.


1 mL serum in no additive (red-top tube).
Minimum quantity: 0.3 mL.
Absolute minimum 0.06 mL.
Ship refrigerated.
Children and adults: ≤0.4 U/mL

Insulin, Free Quest

The free (bioactive) insulin test is recommended for patients who have detectable levels of insulin
autoantibodies.
0.5 mL serum in no additive (red-top tube).
Minimum quantity: 0.3 mL.
Overnight fasting is required. Ship refrigerated.
Children and adults: 1.5–14.9 U/mL.
Insulin levels may vary widely in specimens taken from non-fasting individuals.

Insulin, Serum Quest

0.4 mL serum in no additive (red-top tube only).


Minimum quantity: 0.2 mL.
Absolute minimum 0.1 mL.
Ship refrigerated. Overnight fasting is required. This assay is not recommended for patients with posi-
tive insulin autoantibody. Please use the free insulin assay in those cases. Insulin levels vary widely
in specimens taken non-fasting individuals.
Children: ≤13.7 μU/L.

Islet Cell Antibody Quest

2 mL serum in no additive (red-top tube).


Minimum quantity: 0.5 mL.
Absolute minimum 0.05 mL.
Ship refrigerated.
Children and adults: <1.25 JDF units.
Note: End-point titers are compared with a single international reference standard, and values are
reported in JDF (Juvenile Diabetes Foundation) units.

Leptin, Serum Quest

1 mL serum in red-top or SST tube.


Minimum quantity: 0.2 mL.
Ship refrigerated.
414 16 Laboratory Values for Pediatric Endocrinology

Children
5–9 years 0.6–16.8 ng/mL
10–13 years 1.4–16.5 ng/mL
14–18 years 0.6–24.9 ng/mL

Luteinizing Hormone (LH), Third-Generation Serum Esoterix

Values begin to increase about 2 weeks after birth to a range of 0.02–7.0 mIU/mL within the first 3
months, and then decline to prepubertal values, by the end of the first year.

Range (mIU/mL)
Infants 2 weeks 1 mL serum in no
to 11 months additive (red-top tube).
Minimum quantity:
0.2 mL.
Ship refrigerated.
Prepubertal children 0.02–0.3
12 months to 8 years
Puberty
Tanner stage Age (years) Range Tanner Age Range
(mIU/mL) stage (years) (mIU/mL)
Male Female
1 <9.8 0.02–0.3 1 <9.2 0.02–0.18
2 9.8–14.5 0.2–4.9 2 9.2–13.7 0.02–4.7
3 10.7–15.4 0.2–5.0 3 10.0–14.4 0.10–12.0
4–5 11.8–17.3 0.4–7.0 4–5 10.7–18.6 0.4–11.7
Adults Range (mIU/mL)
Male 1.5–9.0
Female
Follicular 2.0–9.0
Midcycle 18.0–49.0
Luteal 2.0–11.0
Postmenopausal 20.0–70.0

Metanephrine and VMA, Fractionated 24-h Urine Quest

5 mL urine from 24-h urine container.


Minimum quantity: 1.5 mL.
Absolute minimum 1.1 mL.
Collect urine with 25 mL of 6 N HCl to maintain a pH <3. Urine without preservative is acceptable if
pH is <6 and the sample is shipped frozen. Record 24-h urine volume on test-request form and
urine vial. Record patient’s age on test-request form and urine vial. It is preferable for the patient
to be off medications for 3 days before collection. Ship refrigerated.
Metanephrine, Fractionated Random Urine Quest 415

Age
Metanephrine (μg/24 h)
3 months to 4 years 25–117
5–9 years 11–139
10–13 years 51–275
14–17 years 40–189
Normetanephrine, 24 h (μg/24 h)
3 months to 4 years 54–249
5–9 years 31–398
10–13 years 67–503
14–17 years 69–531
Total Metanephrines (μg/24 h)
3 months to 4 years 79–345
5–9 years 49–408
10–13 years 110–714
14–17 years 107–741

Metanephrine, Fractionated Random Urine Quest

Includes metanephrine, normetanephrine, and total metanephrine.


10 mL urine in sterile screw-cap container.
Minimum quantity: 3 mL.
Collect urine with 15 g of boric acid or 15 mL of 6 N HCl to maintain a pH <3. Urine without preser-
vative is acceptable if pH is <6 and the sample is shipped frozen. Record patient’s age on test-
request form and urine vial. It is preferable for the patient to be off medications for 3 days before
collection. Ship refrigerated.

Metanephrine
5–9 years 106–527 μg/g Cr
10–13 years 34–357 μg/g Cr
14–17 years 24–302 μg/g Cr
13–17 years 34–126 μg/g Cr
Normetanephrine
5–9 years 149–781 μg/g Cr
10–13 years 38–523 μg/g Cr
14–17 years 14–302 μg/g Cr
Total metanephrine
5–9 years 255–1,167 μg/g Cr
10–13 years 86–845 μg/g Cr
14–17 years 39–578 μg/g Cr
416 16 Laboratory Values for Pediatric Endocrinology

Metanephrine, Fractionated Free Plasma Quest

Minimum quantity: 1.5 mL.


Metanephrine ≤57 g/mL.
Normetanephrine ≤148 pg/mL.
Metanephrines total ≤205 pg/mL.

Microalbumin

Urine
Range (mg/24 h) Range (mg/g creatinine)
24-h collection <30 <30
Microalbuminuria 30–299 30–299
Clinical albuminuria >300 >300

Osmolality, Random Urine Quest

1 mL urine in sterile screw-cap container.


Minimum quantity: 0.2 mL.
Do not use preservatives. Ship refrigerated.
Children and adults: 50–1,200 mOsm/kg.

Osmolality, Serum Quest

1 mL serum in no additive (red-top tube).


Minimum quantity: 0.2 mL.
Ship refrigerated.
Children and adults: 278–305 mOsm/kg.

Osteocalcin, Serum Quest

This test is performed by using a test kit that has not been approved or cleared by the Food and Drug
Administration (FDA). The performance characteristics of this test have been determined by Quest
Diagnostics, Nichols Institute.
1 mL serum in no additive (red-top tube).
Minimum quantity: 0.5 mL.
Avoid hemolysis. Avoid lipemia. Overnight fasting is preferred. Ship refrigerated.
Parathyroid Hormone (PTH), Intact, Serum Quest 417

Adults
Male Female
9.0–38 ng/mL 8–32 ng/mL
Children
Male Female
5–9 years 47–142 ng/mL 47–142 ng/mL
10–13 years 49–167 ng/mL 49–167 ng/mL
14–17 years 26–203 ng/mL 14–85 ng/mL
Tanner stages
I 20–89 ng/mL 20–89 ng/mL
II 26–91 ng/mL 44–144 ng/mL
III–IV 48–123 ng/mL 31–90 ng/mL

Pancreatic Polypeptide Quest

2 mL EDTA plasma.
0.6 mL minimum.
0.55 mL absolute minimum.
Cord blood.

Term infants <163


Preterm infants <180
Term infants to 6 days <276
1 month for 2 years <644
3–9 years <519
10–13 years <361
14–17 years <297

Parathyroid Hormone Antibody, Serum Quest

1 mL serum in no additive (red-top tube).


Minimum quantity: 0.2 mL.
Absolute minimum 0.125 mL.
Ship refrigerated.
Children and adults: Negative

Parathyroid Hormone (PTH), Intact, Serum Quest

Includes total calcium. The intact PTH is the recommended initial assay for the differential diagnosis
of calcium-related abnormalities.
2 mL serum in no additive (red-top tube).
Minimum quantity: 1 mL.
Absolute minimum 0.275 mL.
Spin and separate serum immediately. Ship refrigerated.
418 16 Laboratory Values for Pediatric Endocrinology

Adults
Males and females 10–65 pg/mL
Children
6–9 years 9–59 pg/mL
10–13 years 11–74 pg/mL
14–17 years 9–69 pg/mL

Parathyroid Hormone and Related Protein (PTH-RP) Quest

0.5 mL plasma in sodium heparin tube at room temperature PTH-RP and releasing factor tube.
Minimum quantity: 0.3 mL.
Absolute minimum 0.25 mL.
Do not thaw. Transfer the plasma to a plastic transport tube and ship frozen.
Males and females: 14–27 pg/mL.

Pregnenolone, Serum Esoterix

2.5 mL serum in no additive (red-top tube).


Minimum quantity: 1.1 mL.
Ship refrigerated.

Adults
Males 13–208 ng/dL
Females 7–188 ng/dL

Eso

Pregnenolone, HPLC-MS/MS
Range (ng/dL)
Premature infants
26–28 weeks, day 4 260–2,104
34–36 weeks, day 4 203–1,024
Infants
1–7 days 150–2,000
Levels decrease after birth, and are within the prepubertal range by 3 months.
Prepubertal children 20–140
Pubertal and adults <151

Progesterone, Serum Esoterix

1 mL serum in no additive (red-top tube).


Minimum quantity: 0.3 mL.
Specify age, sex, and menopausal status on test-request form. Ship refrigerated.
Eso
Proinsulin, Serum Quest 419

Progesterone, HPLC-MS/MS
Range (ng/dL)
Males
1–16 years <10–15
Adults <10–11
Females
1–10 years <10–26
11 years <10–255
12 years <10–856
13 years <10–693
14 years <10–1,204
15 years <10–1,076
16 years <10–1,294
Adult female cycle days
1–6 <10–17
7–12 <10–135
13–15 <10–1,563
16–28 <10–2,555
Postmenopausal <10
Note: Luteal progesterone peaked from 350 to 3,750 ng/dL on
days ranging from 17 to 23.

Progesterone, Free, HPLC-MS/MS

Range (% free) Range (ng/dL)


Males
Adults 1.9–4.2 <0.42
Females
Luteal 1.6–3.3 7.971.5
Follicular 1.5–3.4 <6.1
Postmenopausal 1.6–3.1 <7.1

Proinsulin, Serum Quest

1 mL frozen serum in no additive (red-top tube).


Minimum quantity: 1 mL
0.8 mL minimum.
Overnight fasting is required. Ship refrigerated.
Children and adults: ≤18.8 pmol/L.
420 16 Laboratory Values for Pediatric Endocrinology

Prolactin, Serum Quest

1 mL serum in no additive (red-top tube).


Minimum quantity: 0.5 mL.
Absolute minimum 0.25 mL.
Overnight fasting is preferred. Ship refrigerated.

Adults
Males Females
2.0–18 ng/mL 3–30 ng/mL
Children
Tanner stage Males Females
I ≤10 ng/mL 3.6–12.0 ng/mL
II–III ≤6.1 ng/mL 2.6–18.0 ng/mL
IV–V 2.8–11.0 ng/mL 3.2–20.0 ng/mL

Renin Activity (PRA), Plasma Esoterix

2 mL plasma in EDTA (lavender-top tube).


Minimum quantity: 0.4 mL.
Centrifuge and separate blood at room temperature. Avoid refrigerated temperatures. Patient should
refrain from taking medications, preferably 3 weeks before draw. Patient should be ambulatory for
30 min before draw. Patient should be on a moderate sodium diet during collection. Ship frozen.

Range (ng/mL/h)
Premature
1–7 days 11–167
Full term
1–7 days 2.00–35
Plasma renin activity in newborns is elevated and highly variable.
Children
31 days to 11 months 2.35–37
12 months to 2 years 1.71–11
3–4 years 1.00–6.50
5–9 years 0.50–5.85
10–14 years 0.50–3.30
Range (ng/mL/h)
Adults, normal sodium intake
Upright 0.70–3.30
Supine 0.20–1.60

Sex Hormone-Binding Globulin (SHBG) Esoterix

0.5 mL serum in no additive (red-top tube).


Minimum quantity: 0.2 mL.
0.3 mL absolute minimum.
T3 (Triiodothyronine), Free, Nondialysis Esoterix 421

Specify age and sex on test-request form. Ship refrigerated.


Eso

Range (nmol/L)
Infants
1–23 months 60.0–252.0
Prepubertal children 72.0–220.0
Pubertal children
Males 16.0–100.0
Females 36.0–125.0
Males
20–49 years 16.5–55.9
>49 years 19.3–76.4
Females
20–49 years 24.6–122.0
>49 years 17.3–125.0

Somatostatin Quest

1.8 mL plasma in EDTA (lavender-top tube).


Minimum quantity: 0.6 mL.
0.55 mL absolute minimum.
Draw a prechilled lavender-top tube. Separate and freeze plasma immediately. Do not thaw. Ship
frozen.
<30 pg/mL

T3 (Triiodothyronine), Free, Nondialysis Esoterix

1 mL serum in no additive (red-top tube).


Minimum quantity: 0.5 mL.
Absolute minimum 0.1 mL.
Ship at room temperature.

Range (pg/mL)
Infants
<4 days 2.0–7.9
4 days to 1 month 2.0–5.2
1–11 months 1.6–6.4
Children and adults
1–5 years 2.0–6.0
6–10 years 2.7–5.2
11–19 years 2.3–5.0
≥20 years 2.0–4.4
422 16 Laboratory Values for Pediatric Endocrinology

T3 (Triiodothyronine), Free, Dialysis Esoterix

T3, Free is determined by measuring T3, total, and the dialyzable fraction of labeled T3.
1 mL serum in no additive (red-top tube).
Serum or EDTA plasma ship frozen on dry ice.
Minimum quantity: 0.3 mL.

Age Range (pg/mL) Mean (pg/mL)


Premature (0–4 weeks) 1.35–6.25 3.13
Newborn (0–2 weeks) 0.99–8.03 4.21
Newborn (2 weeks to 12 months) 1.59–6.15 3.87
Prepubertal 1–10 years 2.21–4.99 3.60
Children >10 years and adults 1.81–4.06 2.77
Pregnant females Range (pg/mL) Mean (pg/mL)
First trimester 1.6–3.3 2.5
Second trimester To be determined
Third trimester 1.0–3.2 2.1

T3 (Triiodothyronine), Reverse Esoterix

1 mL serum in no additive (red-top tube).


Separate serum within 45 min of venipuncture; transfer into a plastic transport tube.
Minimum quantity: 0.3 mL.
Ship frozen on dry ice.

Range (ng/dL)
Premature infants
26–31 weeks 33–147
32–35 weeks 49–217
Full-term infants
2–7 days 33–206
8 days to 5 months 13–107
6–12 months 8.1–52.8
Children and adults
1–15 years 8.3–22.9
≥16 years 9.2–24.1

T3 (Triiodothyronine), Total, Radioimmunoassay Esoterix

1 mL serum in no additive (red-top tube).


Separate serum within 1 h; transfer into a plastic transport tube.
Minimum quantity: 0.2 mL.
Ship frozen on dry ice.
T4 (Thyroxine), Free, Nondialysis, Serum Esoterix 423

Range (ng/dL)
Premature infants
26–30 weeks, days 3–4 24–132
Full-term infants
1–3 days 89–405
1 week 91–300
1–11 months 85–250
Prepubertal children 119–218
Pubertal children
11–17 years 80–185
Adults 55–170

T4 (Thyroxine), Free, Direct Dialysis Serum Quest

Direct dialysis is the preferred method for determining Free T4. Free T4 is measured in undiluted
serum by radioimmunoassay of the dialyzable T4.
2 mL serum in no additive (red-top tube).
Minimum quantity: 0.5 mL.
Absolute minimum 0.15 mL.
Ship refrigerated in plastic screw top tube.

Adults
21–87 years 0.8–2.7 ng/dL
Children
Prematures, 25–20 weeks
Birth–7 days 0.5–3.3 ng/dL
Prematures, 31–36 weeks
Birth–7 days 1.3–4.7 ng/dL
>37 weeks 1.2–2.2 ng/dL
Cord blood
0–4 days 2.2–5.3 ng/dL
2 weeks to 2 years 0.8–2.0
3–20 years 1–2.4 ng/dL

T4 (Thyroxine), Free, Nondialysis, Serum Esoterix

0.8 mL serum in no additive (SST tube).


Minimum quantity: 0.3 mL.
Absolute minimum.
Ship frozen.
424 16 Laboratory Values for Pediatric Endocrinology

Range (ng/dL)
Infants
<4 days 0.66–2.71
4 days to 1 month 0.83–3.09
1–11 months 0.48–2.34
Children and adults
1–5 years 0.85–1.75
6–10 years 0.90–1.67
11–19 years 0.93–1.60
20 years 0.82–1.77

T4 (Thyroxine), Total, Serum Esoterix

1 mL serum in no additive (red-top tube).


Minimum 0.2 mL.
Ship refrigerated.

Range (μg/dL)
Premature infants
26–30 weeks, Day 3–4 2.6–14.0
Full-term infants
1–3 days 8.2–19.9
1 week 6.0–15.9
1–11 months 6.1–14.9
Prepubertal children
12 months to 2 years 6.8–13.5
3–10 years 5.5–12.8
Pubertal children
11–17 years 4.9–13.0
Adults 4.2–13.0

Testosterone, Total, Serum Esoterix

0.8 mL serum in no additive (red-top tube).


Minimum quantity: 0.4 mL.
Specify age and sex on test-request form. Ship refrigerated.

Male range Female


(ng/dL) range
(ng/dL)
Premature infants
26–28 weeks, day 4 59–125 5–16
31–35 weeks, day 4 37–198 5–22
Full-term infants 75–400 20–64
Newborns
(continued)
Testosterone Free, Serum or Plasma Esoterix 425

(continued)
Male 1–7 months Levels decrease
rapidly in the first
week to 20–50 ng/
dL, and then
increase to
60–400 ng/dL
(mean = 190)
between 20 and 60
days. Levels then
decline to
prepubertal range of
<2.5–0 by 7 months.
Female 1–7 months Levels decrease
during the first
month to <10 ng/dL
and remain there
until puberty.
Prepubertal children
Males (1–10 years) <2.5–10
Females (1–9 years) <2.5–10
Puberty
Tanner stage Age Range Mean Tanner Age Range Mean
(years) (ng/dL) (ng/dL) stage (years) (ng/dL) (ng/dL)
Male Female
1 <9.8 <2.5–10 4.9 1 <9.2 <2.5–10 4.9
2 9.8–14.5 18–150 42 2 9.2–13.7 7–28 18
3 10.7–15.4 100–320 190 3 10.0–14.4 15–35 25
4 11.8–16.2 200–620 372 4 10.7–15.6 13–32 22
5 12.8–17.3 350–970 546 5 11.8–18.6 20–38 28
Adults 20–50 years Range
(ng/dL)
Male 348–1,197
Female
Premenopausal 10–55
Postmenopausal 7–40

Testosterone Free, Serum or Plasma Esoterix

Includes total, free, and percentage free testosterone.


2 mL serum in no additive (SST tube).
Separate serum within 1 h; transfer into a plastic transport tube.
Minimum quantity: 1 mL.
426 16 Laboratory Values for Pediatric Endocrinology

Testosterone, free, by Blood assays


equilibrium dialysis
Male range (pg/mL) Female range (pg/mL)
Full-term infants
1–15 days 1.5–31 0.5–2.5
1–2 months 3.3–18 0.1–1.3
3–4 months 0.7–14 0.3–1.1
5–6 months 0.4–4.8 0.2–0.6
Prepubertal children
1–10 years 0.15–0.6 0.15–0.6
Puberty Comprehensive values for free testosterone by
dialysis for both males and females throughout
puberty are currently unavailable.
Adults 52–280 1.1–6.3
% Free testosterone
Male range (%) Female range (%)
Full-term infants
1–15 days 0.9–1.7 0.8–1.5
1–2 months 0.4–0.8 0.4–1.1
3–4 months 0.4–1.1 0.5–1.0
5–6 months 0.4–1.0 0.5–0.8
Prepubertal children
1–10 years 0.4–0.9 0.4–0.9
Puberty Comprehensive values for free testosterone by
dialysis for both males and females throughout
puberty are currently unavailable.
Adults 1.5–3.2 0.8–1.4

Testosterone Saliva Esoterix

Testosterone, Saliva, HPLC-MS/MS.

Adults Range (pg/mL)


Male 23–85
Female <9.8

Testosterone Bioavailable Esoterix

Prepubertal Range
children (ng/dL)
Males <0.2–1.3
(1–10 years)
Females <0.2–1.3
(1–9 years)
(continued)
Thyroid-Stimulating Hormone (TSH), Serum Quest 427

(continued)
Puberty
Tanner stage Age Range Mean Tanner Age Range Mean
(years) (ng/dL) (ng/dL) stage (years) (ng/dL) (ng/dL)
Male Female
1 <9.8 <0.2–3.4 1 < 9.2 < 0.2–3.4
2 9.8–14.5 2–58 12 2 9.2–13.7 0.8–4.7 3.6
3 10.7–15.4 12–70 30 3 10.0–14.4 1.1–9.6 4.7
4–5 11.8–17.3 84–350 210 4–5 10.7–18.6 2.3–13.9 6.1
Adults Range (ng/
dL)
Male: 20–39 128–430
years
40–49 years 95–350
50–69 years 95–285
70–79 years 60–240
Female 1.1–14.3

Thyroglobulin Antibody, Serum Quest

1 mL serum in no additive (red-top tube).


Minimum 0.5 mL.
Minimum quantity: 0.15 mL.
Ship refrigerated.
Children and adults: ≤20 IU/mL.

Thyroid Peroxidase Antibody (Anti-TPO), Serum Quest

1 mL serum in no additive (red-top or SST tube).


Minimum 0.5 mL.
Absolute minimum 0.25 mL.
Ship refrigerated.
Children and adults: <35 IU/mL.

Thyroid-Stimulating Hormone (TSH), Serum Quest

2 mL serum in no additive (red-top tube).


Minimum 0.5 mL.
Minimum quantity: 0.5 mL.
Ship refrigerated.

Adults
Males and females (nonpregnant) 0.4–4.5 mU/L
Children
Prematures, 28–36 weeks (first week of life) 0.20–27.90 mU/L
(continued)
428 16 Laboratory Values for Pediatric Endocrinology

(continued)
Cord blood, >37 weeks 1–39 mU/L
Birth to 4 days 3.2–35 mU/L
1–4 weeks 1.0–9.1 mU/L
1–11 months 0.8–8.2 mU/L
1–19 years 0.5–4.3 mU/L

Thyroid-Stimulating Immunoglobulin (TSI) Quest

1 mL serum in no additive (red-top or SST tube).


Minimum quantity: 0.2 mL.
Ship refrigerated.
Children and adults: <140 % of basal activity.

TSH Receptor Antibody (TRAb) Esoterix

1 ml serum in SST tube.


Minimum 0.3 mL.
Ship frozen on dry ice.

Titer at all ages Eso RANGE (U/L)


<1 U/L = Negative
1.1–1.5 U/L = Equivocal
>1.5 U/L = Positive

Thyroxine-Binding Protein/Globulin (TBG), Serum Quest

1 mL serum in no additive (red-top tube) or transport plasma in plastic tube.


Minimum quantity: 0.5 mL.
Ship refrigerated.

Adult male 12.7–25.1 μg/mL


Adult female 13.5–30.9 μg/mL
Pediatric
4–6 Years 14.8–32.9 μg/mL
7–8 Years 16.3–30.7 μg/mL
9–10 Years 15.8–27.4 μg/mL
11 Years 15.5–27.4 μg/mL
12 Years 14.8–26.2 μg/mL
13 Years 13.8–25.2 μg/mL
14 Years 12.2–25.2 μg/mL
15 Years 10.8–23.8 μg/mL
16 Years 10.0–23.8 μg/mL
17 Years 8.5–23.1 μg/mL
Vitamin D, 1,25-Dihydroxy, Serum Esoterix 429

Vanillylmandelic Acid (VMA), Random Urine Quest

10 mL urine from a 24 h collection in sterile screw-cap container.


Minimum quantity: 5 mL.
Absolute minimum 1.1 mL.
Collect urine with 6 N HCl to maintain a pH <6, and the sample is shipped frozen. It is preferable for
the patient to be off medication for 3 days before collection. Ship refrigerated.

Adults
Males and females 1.1–4.1 mg/g Cr
Children
Birth to 6 months 5.5–26 mg/g Cr
7–11 months 6.1–20 mg/g Cr
1–2 years 2.5–21 mg/g Cr
3–8 years 1.7–6.5 mg/g Cr.
9–12 years 1.5–5.1 mg/g Cr
13–17 years 1.5–3.6 mg/g Cr

Vanillylmandelic Acid (VMA), 24-h Urine Quest

10 mL urine from 24-h urine collection.


Minimum quantity: 5 mL.
Absolute minimum 1.1 mL.
Collect urine with 25 mL 6 N HCl to maintain a pH <3. Urine without preservative is acceptable if pH
is <6 and the sample is shipped frozen. Record 24-h urine volume on test-request form and urine
vial. It is preferable for the patient to be off medications for 3 days before collection. Ship
refrigerated.

Adults
Males and females ≤6.0 mg/24 h
Children
3–8 years ≤2.3 mg/24 h
9–12 years ≤3.4 mg/24 h
13–17 years ≤3.9 mg/24 h

Vitamin D, 1,25-Dihydroxy, Serum Esoterix

0.5 mL serum in no additive (red-top or SST tube).


Minimum quantity: 0.3 mL.
Absolute minimum 0.05 mL.
Overnight fasting preferable.
Ship refrigerated.
430 16 Laboratory Values for Pediatric Endocrinology

Vitamin D, 1,25(OH)2, Total


Age Range (pg/mL)
1–9 years 31–87
10–13 years 30–83
14–17 years 19–83
Adult 18–72

Vitamin D, 25-Hydroxy, Serum or Plasma Esoterix

0.5 mL serum in no additive (red-top or SST tube).


Minimum quantity: 0.3 mL.
Absolute minimum 0.05 mL.
Ship refrigerated.

Adults
Males and females 30–100 ng/mL
Deficiency <20
Insufficiency 21–29
Optimum >30
Toxicity >150
S.I. Unit Conversion Table from Esoterix/Labcorp 431

S.I. Unit Conversion Table from Esoterix/Labcorp

Hormone When you know Multiply by To find


ACTH (corticotropin) pmol/L 4.5 pg/mL
Antidiuretic hormone (ADH) pmol/L 1.084 pg/mL
Albumin g/L 0.1 g/dL
Aldosterone, serum nmol/L 36.04 ng/dL
Aldosterone, urine nmol/d 0.3604 μg/24 h
Aldosterone/creatinine nmol/mmol 3.1859 μg/g
Androstanediol nmol/L 29.2 ng/dL
Androstanediol glucuronide nmol/L 46.9 ng/dL
Androstenedione nmol/L 28.64 ng/dL
Androsterone, urine μmol/d 0.2905 mg/24 h
Androsterone/creatinine μmol/mmol 2.568 mg/g
Angiotensin I pmol/L 1.296 pg/mL
Angiotensin II pmol/L 1.046 pg/mL
Angiotensin III pmol/L 0.931 pg/mL
Angiotensin I-converting enzyme U/L 1 mU/mL
Atrial natriuretic peptide (ANP) pmol/L 3.08 pg/mL
C-Peptide nmol/L 3.021 ng/mL
C-Peptide, urine nmol/L 3.021 ng/mL
C-Peptide/creatinine nmol/mmol 26.7109 μg/g
Calcitonin pmol/L 3.418 pg/mL
Calcium mmol/L 4.008 mg/dL
Calcium, urine mmol/d 40.08 mg/24 h
Catecholamines, urine nmol/d 0.1762 μg/24 h

Hormone When you know Multiply by To find


Catecholamines/creatinine nmol/mmol 1.5572 μg/g
Corticosterone nmol/L 34.65 ng/dL
18-Hydroxycorticosterone nmol/L 36.25 ng/dL
Cortisol, serum nmol/L 0.0363 μg/dL
Cortisol, urine nmol/d 0.3625 μg/24 h
Cortisol/creatinine nmol/mmol 3.2045 μg/g
Cortisone nmol/L 0.0361 μg/dL
Creatinine, urine μmol/d 0.1131 mg/24 h
Cyclic Amp, urine μmol/L 1 nmol/mL
Cyclic Amp/creatinine nmol/mmol 0.0088 μmol/g
Dehydroepiandrosterone (DHEA) nmol/L 28.84 ng/dL
Dehydroepiandrosterone-sulfate nmol/L 0.0368 μg/dL
(DHEA-S)
Deoxycorticosterone (DOC) pmol/L 0.0331 ng/dL
18-Hydroxydeoxycorticosterone pmol/L 0.0347 ng/dL
(18-OH-DOC)
11-Desoxycortisol (compound S) nmol/L 34.65 ng/dL
(continued)
432 16 Laboratory Values for Pediatric Endocrinology

(continued)
Hormone When you know Multiply by To find
Dexamethasone nmol/L 39.25 ng/dL
Dihydrotestosterone (DHT) nmol/L 29.04 ng/dL
Dopamine, plasma pmol/L 0.153 pg/mL
Dopamine, urine nmol/d 0.153 μg/24 h
Dopamine/creatinine nmol/mmol 1.3528 μg/g
Endorphin, beta pmol/L 4 pg/mL

Hormone When you know Multiply by To find


Epinephrine, plasma pmol/L 0.1831 pg/mL
Epinephrine, urine nmol/d 0.1831 μg/24 h
Epinephrine/creatinine nmol/mmol 1.6186 μg/g
Estradiol pmol/L 0.272 pg/mL
Estriol pmol/L 0.0288 ng/dL
Estrogens, serum pmol/L 0.271 pg/mL
Estrone pmol/L 0.27 pg/mL
Estrone sulfate pmol/L 0.035 ng/dL
Folic acid pmol/L 0.0441 ng/dL
Follicle-stimulating hormone (FSH) IU/L 1 mIU/mL
Follicle-stimulating hormone, urine IU/d 1 IU/24 h
FSH/creatinine IU/mmol 8.842 IU/g
Gastrin ng/L 1 pg/mL
Glucagon ng/L 1 pg/mL
Growth hormone μg/L 1 ng/mL
Human chorionic gonadotropin (HCG) IU/L 1 mIU/mL
HCG, urine IU/d 1 IU/24 h
HCG/creatinine IU/mmol 8.842 IU/g
5-Hydroxyindoleacetic acid nmol/d 0.1912 μg/24 h
5-HIAA, urine 5-HIAA/creatinine nmol/mmol 1.6906 μg/g
Homovanillic acid (HVA), urine nmol/d 0.1822 μg/24 h
HVA/creatinine nmol/mmol 1.611 μg/g

Hormone When you know Multiply by To find


17-Hydroxycorticosteroids, urine nmol/d 0.3625 μg/24 h
17-Hydroxycorticosteroids/creatinine nmol/mmol 3.2045 μg/g
IGF-I (somatomedin C) nmol/L 7.6490 ng/mL
IGF-II nmol/L 7.5000 ng/mL
Inhibin U/L 0.0010 U/mL
Insulin pmol/L 0.1440 μU/mL
17-Ketosteroids, urine μmol/d 0.2884 mg/24 h
17-Ketosteroids/creatinine μmol/mmol 2.5495 mg/g
Luteinizing hormone (LH) IU/L 1.0000 mIU/mL
Luteinizing hormone, urine IU/d 1.0000 IU/24 h
S.I. Unit Conversion Table from Esoterix/Labcorp 433

Hormone When you know Multiply by To find


LH/creatinine IU/mmol 8.8420 IU/g
Metanephrine, urine nmol/d 0.1972 μg/24 h
Metanephrine/creatinine nmol/mmol 1.7432 μg/g
Metanephrines, total, urine nmol/d 0.1902 μg/24 h
Metanephrines, total/creatinine nmol/mmol 1.6814 μg/g
Methoxytyramine, urine nmol/d 0.1672 μg/24 h
Methoxytyramine/creatinine nmol/mmol 1.4786 μg/g
Norepinephrine, plasma pmol/L 0.1692 pg/mL
Norepinephrine, urine nmol/d 0.1692 μg/24 h
Norepinephrine/creatinine nmol/mmol 1.4957 μg/g
Normetanephrine, urine nmol/d 0.1832 μg/24 h
Normetanephrine/creatinine nmol/mmol 1.6195 μg/g
Osteocalcin nmol/L 6.5000 ng/mL

Hormone When you know Multiply by To find


Parathyroid hormone pmol/L 9.5 pg/mL
Prednisolone nmol/L 36.04 ng/dL
Prednisone nmol/L 35.84 ng/dL
Pregnanediol, urine μmol/d 0.3205 mg/24 h
Pregnanediol/creatinine ng/dL μmol 2.8332 mg/g/mmol
Pregnanetriol, urine μmol/d 0.3365 mg/24 h
Pregnanetriol/creatinine μmol/mmol 2.9747 mg/g
Pregnenolone nmol/L 31.65 ng/dL
17-Hydroxypregnenolone nmol/L 33.25 ng/dL
Progesterone nmol/L 31.45 ng/dL
17-Hydroxyprogesterone nmol/L 33.05 ng/dL
20-Hydroxyprogesterone nmol/L 31.65 ng/dL
Prolactin μg/L 1 ng/mL
Renin (plasma renin activity) ng/L/s 3.6 ng/mL/h
Reverse T-3 pmol/L 0.0651 ng/dL
Secretin pmol/L 3.055 pg/mL
Sex hormone-binding globulin (SHBG) nmol/L 0.0288 μg/dL
(Binding capacity)
Somatostatin-14 pmol/L 1.638 pg/mL
Somatostatin-28 pmol/L 3.276 pg/mL
Testosterone nmol/L 28.84 ng/dL
Free testosterone pmol/L 0.2884 pg/mL

Hormone When you know Multiply by To find


Testosterone, urine nmol/d 0.2884 μg/24 h
Testosterone/creatinine nmol/mmol 2.5495 μg/g
Thyroglobulin μg/L 1 ng/mL
(continued)
434 16 Laboratory Values for Pediatric Endocrinology

(continued)
Hormone When you know Multiply by To find
Thyroid-stimulating hormone (TSH) mU/L 1 U/mL
Thyroxine (T-4) nmol/L 0.0777 μg/dL
Thyroxine-binding globulin mg/L 0.1 mg/dL
Thyrotropin-releasing hormone pmol/L 0.362 pg/mL
(TRH or TRF)
Triiodothyronine (T-3) nmol/L 65.1 ng/dL
Vanillylmandelic acid (VMA), urine nmol/d 0.1982 μg/24 h
VMA/creatinine nmol/mmol 1.7525 μg/g
Vitamin B-12 pmol/L 0.1355 ng/dL
25-Hydroxy-vitamin D nmol/L 0.4006 ng/mL
1,25-Dihydroxy-vitamin D pmol/L 0.4166 pg/mL

Suggested Reading

Laboratory Values from

Endocrinology Expected values & S.I. Unit Conversion Tables. Esoterix/LabCorp, 2014.
Nakamoto J, Mason P. Quest Diagnostics. The quest diagnostics manual endocrinology. San Juan Capistrano, CA:
Quest Diagnostics; 2012.
Index

A B
Abrams, S.A., 147 Bariatric surgery, 21, 143, 351, 355, 356
Acute adrenal insufficiency, 245, 246, 255, 363–365 Baroreceptors, 30, 31
Addison’s disease/primary adrenal failure, 4, 31, 109, Behrman, R.E., 85
117, 134, 173, 210, 240–242, 245, 247–249, 257, Beta cells of the islets of Langerhans, 265, 270
272, 291, 292 Birth history, 11, 12
Adjusted midparental height, 65, 86 Bisphosphonates, 129, 146, 150, 155, 369
Adrenal androgens, 169, 170, 174, 175, 200, 214, 219, Body mass index (BMI), 13–15, 62, 84, 325–327, 403
221–223, 230, 234, 237, 239 in percentiles, 325–329, 346, 348, 352
Adrenal cortex, 233, 238, 240–257 Bone age, 20, 52, 58, 62–64, 67, 69–71, 77, 81, 82,
Adrenal medulla, 233, 257–261, 268, 307, 309 84–88, 106, 108, 109, 111, 116, 117, 169, 171,
Adrenal tumor, 223, 224, 247, 249, 251–252, 255 172, 175, 182, 193, 198–200, 203, 207, 214–216,
Adrenarche, 198, 200, 203, 221, 223, 224, 228, 222–224, 324, 331, 338, 351
237, 252 Bone mineralization, 138, 145, 154
Adrenocorticotropic hormone (ACTH), 1–4, 7, 18, 20, Brannon, P.M., 147
22, 25, 26, 30, 31, 75, 76, 80, 83, 101, 166, 167,
169, 171–174, 200, 207, 220, 223, 233–241,
243–257, 259, 268, 291, 308, 309, 314, 317, 318, C
320, 324, 331, 363, 364, 373, 385, 386, 391, 413 Calcitonin, 100, 119, 123, 124, 127–129, 139, 140, 145,
Adrenoleukodystrophy, 242 151, 152, 156, 369, 377, 387, 414
Age at onset of puberty, 191, 214 human, 377
Aldosterone, 30, 34, 41, 42, 137, 167, 177, 221, 234, Calcium, 4, 75, 78, 107, 115, 119, 123–157, 193, 205,
236, 238, 241, 244, 248, 250, 251, 256–257, 349, 214, 259, 275, 279, 312, 332, 349, 353, 358,
383, 386, 399, 413 367–369, 373, 377, 378, 382, 387–388, 405, 414
Alkaline phosphatase, 129–130, 133, 141, 143–147, 155, Calcium sensing receptor (CASR), 124, 128, 134, 135,
202, 203, 386 137, 149, 150 , 153
Aloia, J.F., 147 Catecholamines, fractionated
Androgen insensitivity, 178–179, 185, 186, 211 24-hr urine, 388–389
Anorchia/testicular regression syndrome, 179, 180, plasma, 389
212–213 Cell membrane GH receptor, 61
Antidiuretic hormone (ADH), 21, 29–31, 35, 41–45, 136, Cell-membrane protein receptors, 4, 243, 267, 269
148, 374, 413 Central diabetes insipidus (DI), 29, 32–41, 76, 206, 379
Anti-Müllerian hormone (AMH), 162, 166, 168, 176, Central nervous system control of appetite, 329–332
178, 180, 183, 184, 194, 201, 212, 223, 230, 387 Central nervous system tumors, 19–21, 78, 202,
5α-reductase deficiency, 167 204–206, 227
Arginine vasopressin (AVP), 20, 29, 30, 33–35, 39, 43, Central (complete or true) precocious puberty (CPP),
45, 198, 378, 387 22, 24, 169, 190, 193, 194, 197, 206, 217–220,
Arm span, 14, 15, 52, 84, 87, 211 222, 224
Atrial natriuretic hormone (ANH), 387 Cerebral edema (CE), 32, 276–277, 279, 366, 380
Autocrine, 1, 2 Cerebral salt wasting (CSW), 34, 42
Autoimmune polyendocrinopathy syndrome, 100, 133, Cholecalciferol, 127, 128, 138, 146, 150, 151
134, 240, 272 Circadian rhythm, 21, 80, 239
Autoimmune polyglandular endocrinopathy, 240–242 Clinton, S.K., 147
Autoimmune polyglandular syndrome, 240–242, 246, 291 Comorbidities of obesity, 339–352, 356

© Springer International Publishing Switzerland 2016 435


D.M. Styne, Pediatric Endocrinology, DOI 10.1007/978-3-319-18371-8
436 Index

Congenital adrenal hyperplasia (CAH), 64, 68, 166–171, E


173–174, 176, 181–185, 198, 217, 219–221, 223, Ectopic hormone production, 390
230, 233, 235, 237–240, 247–249, 256, 314, 363, Endemic cretinism, 102, 104, 110
373, 385, 387 Endocrine, 1–8, 11–15, 17–19, 21–25, 35, 47, 58–62, 65,
Congenital adrenal hypoplasia (AHC), 192, 205, 242 67, 74–86, 91, 109, 116, 118, 124, 127, 138, 140,
Congenital hypothyroidism, 64, 83, 96, 98, 101–108, 111, 149, 150, 152, 153, 159, 165, 168, 170, 179, 180,
112, 131 184, 187, 191–202, 205, 207, 210, 213, 214,
Constitutional delay in growth, 63, 68–70, 84, 192, 216–218, 220–228, 230, 233, 250–252, 258, 260,
203, 215 267–269, 272, 275, 290, 309, 314, 332, 337–339,
Constitutional delay in puberty (CDP), 194, 201–203, 213, 351, 356, 363–376, 385
214, 218 Endocrine causes of obesity, 338
Constitutive activation, 5, 116 Energy expenditure, 208, 291, 336, 338, 352–354
Corticotropin (or ACTH), 1, 18, 19, 30, 234, 235 Energy intake, 208
Corticotropin-releasing factor (CRF), 1, 3, 4, 18, Epigenetics, 72, 77, 330, 337, 360
233–235, 241, 244, 245, 249–251, 254 Ergocalciferol, 127, 128, 141, 146, 150, 151, 378–379
Cortisol, 1–3, 7, 8, 19, 41, 75, 80, 83, 86, 166, 167, 170, Estradiol, 60, 61, 82, 88, 124, 176–178, 191, 194, 195,
171, 174, 177, 182, 199, 221, 234–241, 244–257, 198–201, 216, 220–222, 225, 227, 233, 394–395
266, 270, 291, 306–310, 314, 318–321, 324, 344, Estradiol, serum, 220, 222, 395
364, 371, 380, 390–391, 414 Euthyroid sick syndrome, 111
salivary, 391 External genitalia, 163, 165, 166, 168, 175, 179–181,
Cortisol-binding globulin (CBG/transcortin), 237 185, 209, 210, 221, 222
Cortisol serum total LC/MS/MS, 391 Extracellular domain, 4, 5
Cortisol urinary free 24-hour, 390
C peptide, 204, 265, 274, 300, 301, 308, 317, 319, 321,
371, 387, 414 F
Craniopharyngioma, 20–21, 33, 35, 44, 45, 78, 85, 192, Familial hypocalciuric hypercalcemia (FHH),
205, 217, 339 124, 148–150, 153
Cushing disease, 4, 8, 192, 237, 239, 249–256, 267, 364 Familial short stature, 82
Cushing’s syndrome, 2, 7, 64, 83, 86, 221, 222, 229, 237, Family history, 12, 13, 84, 109, 131, 153, 154, 203, 213,
247, 249, 251–257, 338, 339, 349, 351 216, 219, 220, 223, 230, 273, 293, 299, 346, 347,
Cytokine receptors, 4, 18 349, 367, 369
Cytoplasmic steroid receptors, 6 Fetal adrenal gland, 233
Fine needle aspiration under ultrasound guidance, 118–119
Fisher, D.A., 133, 142, 148
D Follicle-stimulating hormone (FSH), 4, 18, 25, 26, 76,
D-arginine-D-amino-vasopressin (DDAVP), 38–40, 375, 184, 191–195, 197–201, 205, 206, 209, 211, 212,
378, 379 214, 219, 220, 222, 224, 227, 230, 396, 414
Defects in gluconeogenesis, 314–316, 321–322, third generation, serum, 396
324, 372 Free FT4 (or FT4), 93, 97–98, 101, 103, 105, 109, 110,
Dehydroepiandrosterone sulfate (DHEAS), 174, 177, 183, 113, 116
191, 200, 203, 221, 223, 228, 233, 238, 239, 414
Delayed puberty, 14, 15, 21–25, 52, 62, 67, 69, 75, 77,
81, 82, 84, 192, 201–207, 213–219, 228, G
249, 295 Gallagher, J.C., 147
Developmental defects of the midline, 24 Gallo, R.L., 147
DHEA Serum, 392–393 Gardner, D.G., 86
DHEA Sulfate, serum, 393 Gastrointestinal (GI) hormones, 267–270
Diabetic ketoacidosis (DKA), 40, 43, 116, 125, 266–267, GeneTests, 8, 182–184
270, 272–281, 284, 287, 289–291, 295, 296, 300, Genetic factors indiabetes, 271
303, 365–366, 370, 373, 382 Genotype, 159–161, 210
Dietary history, 280–281, 317, 323, 347, 348 Germinoma, 21–22, 33, 39, 78, 85, 192, 206, 217,
Dihydrotestosterone (DHT), 163–167, 176–178, 183, 219–221
184, 198–200, 230, 394, 414 GH-binding protein (GHBP), 4, 6, 62, 63, 76–77, 398
1,25-Dihydroxy vitamin D, 123, 125–128, 131, 132, 137, Glucagon, 4, 5, 58, 266–271, 281, 290–291, 307, 311,
139, 143–145, 144, 145, 147, 152, 155, 413 314, 318, 322, 323, 330, 356, 372, 376, 397, 414
Disorders of sexual differentiation (DSD), 159–187, 201, Glucagon Hcl, 379
208–211 Glucocorticoid withdrawal, 255
Diurnal rhythm, 7, 97, 197, 253 Glucorticoid preparations, 246, 379, 380
Durazo-Arvizu, R.A., 147 Glucose infusion rate, 305, 308, 311, 313, 372
Dyslipidemia, 13, 228, 230, 294, 295, 344, 346–348, 356 Glucose regulatory factors, 306–307
Index 437

GLUT 4, 266, 270, 306 Hypercalcemia, 36, 124, 126–130, 135, 137, 138, 146,
Glutamic acid decarboxylase 65 (GAD 65) antibodies, 148–156, 259, 368–369, 381
264, 271, 272, 274 Hyperglycemia, 34, 43, 264–267, 272, 273, 279, 280,
Glycogen storage disease, 308, 310, 318, 323, 372 283, 288–290, 294, 296–300, 302, 303, 309, 311,
Glycoprotein hormones, 18, 205 312, 317, 374
Glycosylated hemoglobin/hemoglobin A1c, 288 Hypergonadotropic hypogonadism, 73, 134, 177, 192,
Goiter, 14, 15, 98–102, 104–106, 108–110, 112–114, 197, 203, 207–213
117, 370 Hyperinsulinism, 63, 88, 259, 309–311, 313, 314,
Gonadal DSD, 179–180 316–317, 321–324, 331, 338, 372, 373
Gonadal sex, 159, 161–162 Hyperkalemia, 128, 138, 149, 167, 168, 171, 172, 184,
Gonadarche, 194–200, 203, 214 238, 245, 248, 277, 279, 363–366, 373–374
Gonadotropin-releasing hormone (GnRH), 4, 18, 22, 82, Hypernatremia due to diabetes insipidus, 39, 374–375
89, 169, 193–197, 199, 201, 204, 206, 212–213, Hyperosmolality, 32, 323, 372
215, 217, 219, 220, 222, 225, 227, 228 Hyperparathyroidism, 7, 130, 143–145, 148–153, 155,
Gonadotropin-releasing hormone analogues (GnRH-As), 259, 269, 369
224–225 Hyperphosphatemia, 84, 125, 127, 130, 131, 136–138,
G protein complex, 4, 5 143, 151, 367
Graves’ disease, 91, 92, 97–99, 105, 109, 112–114, 116, Hypertension, 13, 34, 71, 96, 113, 117, 148, 149, 167,
117, 382 174, 177, 209, 210, 216, 228, 230, 249, 256–260,
Greenspan, F.S., 86 277, 292, 295, 296, 331, 343, 347–350, 352, 356,
Greulich, W.W., 58, 69 369, 375, 376, 379, 382, 383
Growth charts, 13, 47, 48, 50–52, 55, 56, 65–67, 69, 70, Hypertension due to pheochromocytoma, 375–376
74, 75, 203, 328 Hyperthyroidism, 74, 85, 92, 93, 97–99, 101, 105, 106,
Growth factors, 1, 62, 66, 67, 175, 204 109, 110, 112–117, 120, 148, 150, 152, 221, 241,
Growth hormone (GH), 4–6, 17–27, 47, 56, 58–70, 269, 370, 371, 381
72–89, 102, 124, 127, 140, 145, 172, 193, 203, Hyperthyroidism in the neonate, 370
206–207, 209, 211, 215, 216, 230, 240, 243, 252, Hypocalcemia, 84, 115, 123–126, 128–143, 151, 155,
259, 266–268, 270, 291, 307–310, 314, 317–320, 296, 366–368, 377, 378, 380, 381
321, 322, 324, 330, 338–339, 351, 371, 379, Hypoglycemia, 6, 12, 26, 61, 101, 168, 207, 240, 265,
397–398, 414 305, 338, 363, 379
deficiency, 17, 19, 20, 23–25, 27, 33, 56, 58–60, Hypogonadotropic hypogonadism (HH), 19, 25, 27, 73,
62–65, 67, 68, 74–83, 86, 179, 206, 215, 216, 78, 185, 192, 194–196, 203–207, 213, 214, 216,
250, 256, 308, 314, 324, 338–339, 351, 382, 398 218, 242, 331
resistance, 59, 62, 68, 76–78, 80, 317 Hyponatremia, 30, 34, 40–44, 75, 167, 172, 173, 184,
secretion tests, 18, 22, 58–64, 70, 74, 77–80, 83, 86, 238, 245, 248, 323, 363–365, 372–374
88, 89, 259, 267, 268, 338 Hyponatremia due to the syndrome of inappropriate
serum, 397–398 ADH secretion, 45
Growth hormone release inhibitory factor (or Hyponatremia/hyperkalemia due to adrenal insufficiency,
somatostatin or SRIF or SS), 58 373–374
Growth hormone releasing factor (GHRH/GRF), 4, 17–19, Hypoosmolality, 32
58, 59, 76, 80, 84, 136 Hypoparathyroidism, 7, 109, 119, 131–137, 149, 240,
Growth velocity, 52, 56, 70, 87, 193, 233 242, 367–369, 377, 379
Gut microbiome, 336, 339, 350, 357 Hypophosphatemic rickets, 75, 84, 125, 142,
Gynecomastia, 177, 178, 211, 217, 221, 223, 252 145–146, 221
Hypophysiotropic hormones, 1
Hypophysiotropic portal system, 59, 193, 234
H Hypothalamic inhibiting factors, 17
Hamartomas of the tuber cinereum, 217, 219, 224 Hypothalamic pituitary axis, 4, 17–27, 91, 199, 218, 219,
Hashimoto’s thyroiditis, 71, 98–100, 109–110, 112–114, 226, 235, 249, 251
209, 246, 272, 373 Hypothalamic releasing factors, 17
Health disparities, 325 Hypothalamic thyrotropin-releasing hormone
Hemoglobin A1c (HgbA1c), blood, 230, 274, 286, 288, (TRF), 91
292, 294, 296, 300, 352, 398 Hypothalamus, 1, 2, 17, 19, 23, 24, 29, 30, 32, 44, 58,
Home measurement by glucometer, 281–282 59, 61, 91, 96, 101, 193, 195, 197, 198, 204, 219,
Homeodomain proteins, 17, 25 233–235, 237, 243, 330, 332
Honeymoon period, 273, 289, 292, 300, 303 Hypothyroidism, 15, 20, 24, 31, 34, 52, 56, 62, 64, 67,
Hormone-receptor complex, 6, 7 68, 74–76, 78, 80, 83, 85, 91, 92, 96–113, 115,
Hungry bone syndrome, 115, 125, 128, 149 116, 119, 131, 134, 192, 205, 208, 217, 219,
25 Hydroxy vitamin D, 126, 128, 139, 141, 144–146, 223, 240–243, 247, 292, 338, 348, 351, 352,
149, 415 365, 370
438 Index

I Loss of function mutations of MKRN3, 219


IA-2 (islet cell associated phosphorylase) antibodies, Luteinizing hormone (LH), 4, 18, 26, 76, 164, 179,
271, 272 192–194, 205, 242, 403, 414, 415
Iatrogenic glucocorticoid deficiency, 247
Iatrogenic obesity, 339
Idiopathic hypopituitarism, 25–27, 206–207 M
Idiopathic short stature, 67, 69 Magnesium, 123, 125–126, 131–133, 136–137, 257, 275,
IGF-BINDING PROTEIN-3 (IGFBP-3), 63, 401–402 279, 314, 365, 367, 368, 380
Inhibin B, 180, 184, 201, 203, 212–214, 223, 402 Manson, J.E., 147
Insulin, 5, 21, 40, 58, 109, 125, 193, 239, 263, 305, 330, Maternal age of menarche, 12, 13, 84
366, 379, 402 Maturity-onset diabetes of the young (MODY), 264, 265,
preparations, 282, 283, 380 273, 298, 301–302
pump, 275, 276, 281, 286–289, 291, 303 Mayne, S.T., 147
receptor, 5, 6, 63, 65, 266, 316 McCune–Albright syndrome, 5, 88, 112, 116, 130, 146,
resistance, 82, 228, 249, 264, 268, 270, 286, 289, 217, 221, 223, 225, 252
296, 299, 316, 317, 330, 331, 337, 339–346, 348, Measurement of growth, 47–58
352, 355 MECASERMIM (rhIGF-1), 77
Insulin-like growth factor 1 (IGF-1), 5, 6, 59, 62, 63, 77, Medical history, 11–13, 84, 181, 299, 309, 319
78, 88, 127, 259, 338, 400–401 Medication and complementary medical
Insulin like growth factor binding protein 3 (IGFBP3), history, 11
63, 77, 85 Medications for pediatric endocrinology, 377–383
Insulin-like growth factor 1 receptors (IGF1R), 6 Medullary carcinoma of the thyroid (MCT), 100, 118,
Insulin-receptor substrates (IRSs), 5 119, 128, 260
Internal sexual ducts, 162–164 Membrane protein receptor, 124, 126, 221, 235, 243,
Intracellular domain, 4 267, 269, 307
Intracrine, 1, 2, 230 Metabolic syndrome, 228, 230, 299, 329, 337,
Ionized calcium, 123, 131, 135, 149, 150, 155, 368 339–344, 350
Islet cell antibodies, 264, 300, 402 Metformin, 230, 300, 350, 355, 356, 381
Microphallus, 75, 76, 105, 165, 179, 186, 194, 203, 207,
314, 319, 363, 371, 372
J Microvascular and macrovascular complications of
Janus kinase-signal transducer and activator of diabetes, 294
transcription (Jak-STAT) pathway, 62 Mineralocorticoids, 124, 168, 172, 174, 176, 177, 199,
Jones, G., 147 221, 233, 234, 238, 240, 242–247, 249, 252–253,
255–257, 363, 364, 373, 374, 380
Motivational interviewing, 353
K Multidisciplinary team management, 186
Kallmann syndrome (KS), 192, 203–205 Multiple-dose injection regimen, 283–286
Ketotic hypoglycemia, 308, 315, 317–319, 321–323, 372 Multiple-dose injections, 283–286, 289, 300
Kinases, 4, 5, 62, 65, 118, 205, 242, 252, 300, 318, Multiple endocrine neoplasia, 22, 23, 88, 100, 118, 149,
347, 355 152, 153, 252, 258–260, 267–269, 314, 369
Kisspeptin, 194, 197, 204, 218–219
Klinefelter’s syndrome, 15, 52, 86, 88, 110, 160, 179,
180, 192, 201, 203, 210–211, 213, 223 N
Kovacs, C.S., 147 Natruretic peptides, 30, 32, 33
Negative feedback, 3, 19, 59, 127, 178, 194–196, 199,
235, 240, 251
L Negative feedback loop, 127
Laboratory values for pediatric endocrinology, Neonatal diabetes mellitus (NDM), 265, 297–299
385–415 Neonatal hyperinsulinemic hypoglycemia, 268
Langerhans cell histiocytosis, 23, 33, 68, 110 Neonatal hypoglycemia, 26, 88, 101, 311, 314, 317
Lengthening of the QTc interval, 132, 134 Neonatal screening, 83, 101, 103, 105, 106, 168, 170
Leptin, 4, 199, 268, 330–332, 339 Nephrogenic diabetes insipidus, 29, 32, 34–36, 39, 41
Leptin, serum, 403 Neuroblastoma, 249, 258, 260–261, 268
Leydig cells, 5, 162, 176, 179, 180, 183, 184, 194, Neurophysin, 19, 29, 39
198, 199, 205, 210–212, 217, 220, 221, 223–227, Nodular adrenal hyperplasia, 251–252, 255, 267
257, 317 Nonalcoholic liver disease (NAFLD), 350
Lifestyle modification, 230, 347, 349, 350, 352–355, 357 Nonketotic hyperosmolar coma, 303
Lipid (prostaglandin) structures, 3, 331 Normal bone growth, 138–140
Index 439

O Primary IGF deficiency, 68


Obesity, 12, 21, 58, 108, 136, 189, 243, 263, 316, 325 Proinsulin, 265, 270, 298, 331, 406–407
Obesogenic environment, 337–338 Prolactin, serum, 407
Online mendelian inheritance in man, 8, 156 Pro-opiomelanocortin (POMC), 235, 243, 330–331
Osmolality, 30, 32, 34, 36–39, 41–43, 80, 278, 323, 372, Propranolol, 80, 115, 116, 259, 311, 319, 370, 371,
375, 404 375, 382
Osmolality, serum, 29–33, 36–38, 41, 43, 245, 278, 303, Protein binding, 93–94, 96, 123, 132, 201, 239
374, 375, 405 Pseudohypoparathyroidism (PHO), 2, 68, 79, 83–84,
Osmoreceptor, 30 131, 133, 135–136, 138, 143, 338, 351, 367, 379
Osteogenesis imperfecta, 68, 140, 155, 156, 377, 382 Psychogenic polydipsia, 29, 32, 34, 36–39
Osteoporosis, 88, 141, 153–156, 193, 207, 249 Psychosocial dwarfism, 13, 65, 68, 78–81, 84
Overweight, 218, 264, 325, 329, 338, 340, 353, 354, 357, Pubertal growth spurt, 60, 63, 64, 193, 202–204
358, 360 Pyle, L., 58, 69
Ovotesticular DSD, 162, 163, 179–180
Ovulation and menarche, 201–202
R
Radiation damage to the gonads, 212
P Radiation damage to the hypothalamus, 24, 332
Paracrine, 1, 2, 62, 65, 127, 162, 168 Radioactive iodine (131I; RAI) therapy, 99, 105, 112,
Paragangliomas, 258, 268 115–117, 120, 370
Parathyroid hormone (PTH), 2, 4, 7, 83, 123–128, 130–136, Radioiodine imaging of a thyroid gland, 97
138, 141–144, 147–151, 153, 155, 259, 369, 405 Renal osteodystrophy, 74, 143
Parathyroid hormone-and related protein (PTH-RP), Renal water channel aquaporin, 35
405–406 Renin activity (PRA), plasma, 93, 96, 98, 105,
Parathyroid hormone (PTH), intact, 126, 131, 405 111, 409
Parathyroid hormone-related protein (PTHrP), 123, 126, Reverse T3 (RT3), 93, 96, 98, 105, 110, 111
127, 148, 150, 369 Rickets, 15, 68, 75, 84, 129, 130, 141, 143–147,
Pediatric Sex steroid and gonadotropin and their assays, 154–156, 221, 379
200–201 Rocaltrol, 368
Peptide hormones, 4, 5, 58, 128, 212, 235, 269, 330 Rosen, C.J., 147
Peripheral Precocious Puberty, 206, 218–221, 224, 226 Ross, A.C., 147
Phenotypic sex, 159, 162–165
Pheochromocytoma, 119, 152, 249, 252, 258–261, 268,
269, 349, 375–376 S
Phosphate, 124–126, 129–131, 135, 138, 140, 143–146, Secondary adrenal insufficiency, 244, 245, 255, 331
149, 151–153, 155, 157, 275, 279, 369, 381–382 Secondary endocrine disorders, 4, 207
Phosphorylation, 4, 5 Secondary hypothyroidism, 91, 98, 105
Physical examination, 11, 13–15, 47, 84, 85, 89, 99, 101, Secondary sexual development, 176, 191, 210, 214–216
131, 148, 155, 156, 213, 292, 319 Secular trend, 191
Pituitary adenomas, 3, 22–24, 250, 252, 259, 268 Seminiferous tubules, 194, 205, 210, 220, 224
Pituitary gland, 1, 2, 17, 19, 20, 22, 25, 30, 59, 78, 86, Sense of thirst, 21, 30, 32, 33
91, 105, 108, 116, 193–195, 205, 234, 240, 243, 7 Trans-membrane domain G protein coupled type 1
250, 251, 255, 267, 268, 330, 363 parathyroid hormone receptor (PTHR), 126
Pituitary stalk section, 19, 29 Seven transmembrane domains, 4, 5, 92, 126, 220
Polycystic ovarian disease, 169, 192, 223 Sexual differentiation, 159–187, 194, 201, 208–211
Polycystic ovarian syndrome (PCOS), 169, 174, 199, Sexual precocity, 22, 86, 169, 217–218, 221–222, 227
201, 202, 223, 228–230, 299, 316, 344 Shapses, S.A., 147
Positive feedback, 2, 201–202 Sick day management, 289–290
Posterior pituitary gland or neurohypophysis, 19, 25, Signal transducer and activator of transcription (STAT),
29–45 4, 6, 62
Premature Adrenarche, 200, 217, 221, 223, 228, 252 Small for gestational age (SGA), 69–73, 77, 81, 130, 223,
Premature Leydig and germinal cell maturation, 198, 296, 298, 309, 310, 317, 319, 337, 346, 367, 383
220, 227 Social gender, 166
Premature Thelarche, 217, 222, 224 Somatropin (Growth hormome), 382
Prenatal history, 12 SRY gene, 160–162, 179, 180, 184, 211
Prevention of obesity, 301 Stadiometer, 14, 49
Primary endocrine disorders, 4, 207 STAT. See Signal transducer and activator of
Primary hyperaldosteronism, 256–257 transcription (STAT)
Primary hypothyroidism, 64, 75, 91, 92, 97, 98, 105–110, Stature in percentiles or standard deviations, 51, 58,
208, 217, 223 65–67, 154, 155, 200, 212, 325
440 Index

Steroid, 1–4, 6, 7, 24, 60, 63, 64, 66, 78, 94, 95, 127, Thyroxine (Levo), 380
128, 152, 155, 162, 163, 165, 170, 172–178, Thyroxine (or T4), 82, 92–97, 100, 101, 103,
181–183, 194–203, 206, 208, 210, 214, 215, 105–107, 110–114, 116, 118, 119, 293, 352,
217–221, 224, 226, 227, 233, 235, 236, 239–240, 370, 371, 409
247–248, 252, 257, 344, 364 Triiodothyronine (or T3), 92–94, 117, 415
Styne, D.M., 68, 86 Triphasic Response after Surgery for
Syndrome of inappropriate secretion of Antidiuretic Craniopharyngioma, 40, 44, 45
Hormone (SIADH), 31, 32, 34, 40–44, 374 T3 (triiodothyronine), total, 408
T4 (thyroxine), total, 409
Turner syndrome, 51, 68, 71–72, 81, 82, 110, 159, 162,
T 180, 192, 194, 201, 203, 208, 209, 211, 213, 216,
Tall stature, 83, 86–89, 175, 222, 338, 351 301, 383
Tanner staging or Sexual maturation ratings (SMR), 189, Type 1 diabetes mellitus, 35, 74, 82, 85, 109, 134,
223, 386, 393–396, 400, 403, 405, 407, 410, 411 240, 241, 264, 269–273, 280–294, 297–298, 307,
Tertiary adrenal insufficiency, 238, 243, 245 316, 348
Tertiary endocrine disorders, 4 Type 2 diabetes mellitus, 82, 264, 299–303, 328, 340,
Tertiary hypothyroidism, 91, 92, 98, 101, 105, 111 348, 356
Testicular enzyme deficiencies, 176
Testosterone, 3, 11, 18, 64, 81, 82, 87, 140, 160,
162–165, 167, 172, 176–179, 183, 184, 186, 191, U
193–196, 198–201, 205, 207, 210–217, 220, 223, Ultrasensitive TSH assays, 97, 98
226, 227, 230, 238, 239, 252, 383, 410–411, 415 Ultrasonographic scanning of the thyroid gland, 98, 99,
bioavailable, 411 110, 117
total, 410 Upper-to-lower segment ratio, 14, 15, 52, 58, 62, 64, 84,
Tetany, 124, 130–132, 134, 137, 145, 156, 241, 257, 87, 103, 109, 204, 211
366, 367
T3 (triiodothyronine), free, 408
T4 (thyroxine), free, 409 V
The built environment, 31 Vasopressin, 4, 17–21, 25, 26, 29–45, 198, 199, 234, 238,
The critical sample, 309, 316, 320–322, 371 245, 374, 378, 383, 387
The fasting state, 267, 295, 307, 322, 346 Vitamin D deficiency, 127, 128, 130, 133, 140–149,
The fed states, 266, 305, 307, 313, 319, 322 378–379
Thyroglossal duct cyst, 96, 110 Vitamin D-dependency rickets, 142, 144, 379
Thyroid antibodies, 106, 109, 111 Vitamin D,25-hydroxy, serum/plasma esoterix, 413
Thyroid-binding globulin (TBG), 93, 96, 97, 101, 105, Vitamin D-resistant rickets, 68, 123, 142, 144, 379
111, 113, 412
Thyroid carcinoma, 98, 99, 110, 117–120, 150, 252,
269, 356 W
Thyroidectomy, 115, 118, 119, 260 Weight in percentiles/standard deviations, 14, 278,
Thyroid hormone receptors, 6, 94, 105 328, 356
Thyroiditis, 71, 98–100, 109–110, 116, 120, 241, 293 Weight loss and chronic disease, 207–208
Thyroid nodules, 117, 118
Thyroid peroxidase antibody (Anti-TPO), serum, 412
Thyroid stimulating hormone (TSH), 4, 18, 20, 25, 26, X
58, 64, 76, 91, 92, 94, 96–113, 116, 118–120, 46 XX DSD, 166, 169, 171, 174, 175, 177, 182, 185, 210
131, 136, 206–208, 219, 240–241, 243, 293, 338, 46 XY DSD, 162, 166, 176–179, 185, 186, 211
352, 370, 412, 415
Thyroid-stimulating hormone (TSH), serum, 412
Thyroid-stimulating immunoglobulin (TSI), 92, 113, 412 Z
Thyroid storm, 115–117, 370–373, 382 ZnT8 (zinc transporter molecules) antibodies, 273, 274

You might also like