Annals of Clinical Biochemistry: An international journal of biochemistry and laboratory medicine http://acb.sagepub.com/

17-Hydroxyprogesterone in children, adolescents and adults John W Honour Ann Clin Biochem 2014 51: 424 originally published online 7 April 2014 DOI: 10.1177/0004563214529748 The online version of this article can be found at: http://acb.sagepub.com/content/51/4/424

Published by: http://www.sagepublications.com

On behalf of: Association for Clinical Biochemistry and Laboratory Medicine

Netherlands Society for Clinical Chemistry and Laboratory Japan Society of Clinical Chemistry

Additional services and information for Annals of Clinical Biochemistry: An international journal of biochemistry and laboratory medicine can be found at: Email Alerts: http://acb.sagepub.com/cgi/alerts Subscriptions: http://acb.sagepub.com/subscriptions Reprints: http://www.sagepub.com/journalsReprints.nav Permissions: http://www.sagepub.com/journalsPermissions.nav

>> Version of Record - Jun 11, 2014 OnlineFirst Version of Record - Apr 7, 2014 What is This?

Downloaded from acb.sagepub.com at UNIV TORONTO on July 2, 2014

Review Article Annals of Clinical Biochemistry 2014, Vol. 51(4) 424–440 ! The Author(s) 2014 Reprints and permissions: sagepub.co.uk/journalsPermissions.nav DOI: 10.1177/0004563214529748 acb.sagepub.com

17-Hydroxyprogesterone in children, adolescents and adults John W Honour

Abstract 17-Hydroxyprogesterone (17-OHP) is an intermediate steroid in the adrenal biosynthetic pathway from cholesterol to cortisol and is the substrate for steroid 21-hydroxylase. An inherited deficiency of 21-hydroxylase leads to greatly increased serum concentrations of 17-OHP, while the absence of cortisol synthesis causes an increase in adrenocorticotrophic hormone. The classical congenital adrenal hyperplasia (CAH) presents usually with virilisation of a girl at birth. Affected boys and girls can have renal salt loss within a few days if aldosterone production is also compromised. Diagnosis can be delayed in boys. A non-classical form of congenital adrenal hyperplasia (NC-CAH) presents later in life usually with androgen excess. Moderately raised or normal 17-OHP concentrations can be seen basally but, if normal and clinical suspicion is high, an ACTH stimulation test will show 17-OHP concentrations (typically >30 nmol/L) above the normal response. NC-CAH is more likely to be detected clinically in females and may be asymptomatic particularly in males until families are investigated. The prevalence of NC-CAH in women with androgen excess can be up to 9% according to ethnic background and genotype. Mutations in the 21-hydroxylase genes in NC-CAH can be found that have less deleterious effects on enzyme activity. Other less-common defects in enzymes of cortisol synthesis can be associated with moderately elevated 17-OHP. Precocious puberty, acne, hirsutism and subfertility are the commonest features of hyperandrogenism. 17-OHP is a diagnostic marker for CAH but opinions differ on the role of 17OHP or androstenedione in monitoring treatment with renin in the salt losing form. This review considers the utility of 17-OHP measurements in children, adolescents and adults.

Keywords Steroid hormones, evaluation of new methods, endocrinology Accepted: 5th March 2014

Introduction 17-Hydroxyprogesterone (17-OHP) in serum is a critical diagnostic test for congenital adrenal hyperplasia (CAH) due to 21-hydroxylase deficiency which is amongst the commonest genetic disorders in man. The clinical features of CAH have been known for some time, even as far back as 1865, in an autopsy account of a patient described by Luigi de Crecchio. The basis for the problems could not be ascribed at that time. Bongiovanni published a partial translation

of the original account.1 ‘The cadaver of a ‘‘man’’ having a large penis with first degree hypospadias and without externally visable or palpable gonads. On dissection a vagina, uterus, fallopian tubes and ovaries were found within the abdomen’. Unusual steroids in the urine of Institute of Women’s Health, University College London, London, UK Corresponding author: John W Honour, Institute of Women’s Health, University College London, 74 Hunter Street, London WC1E 6AU, UK. Email: [email protected]

Downloaded from acb.sagepub.com at UNIV TORONTO on July 2, 2014

Honour

425

such patients with adrenal virilism and ambiguous genitalia were first chemically characterised in 1937.2 The nature of the steroid hormones and their link with cholesterol, sex steroids, cortisol and aldosterone became apparent over the next 20 years.3 A biosynthetic pathway was defined and generally accepted, although deviations to the route are possible.4 Prior to 1968, excess androgen production in CAH was defined by analysis of 17-ketosteroids (androgens) in urine, an obsolete colorimetric test since specific steroids can now be measured in blood. Pregnanetriol was known to be elevated in urine of patients with virilism before this was accepted as the main metabolite of 17-OHP. CAH due to 21-hydroxylase deficiency was first accepted in 19555 and now constitutes a spectrum of disease with varying severity and biochemical features consistent with expression of mutations in the CYP21A2 gene. The laboratory often gets requests for 17-OHP during investigations of patients with androgen excess.6 This review addresses the utility of measuring 17-OHP concentrations when investigating children, adolescent and adult patients with androgen excess. The information herein will help a clinical biochemist answer questions raised by clinicians. The scientist can help with interpreting steroid/biochemical data and suggest protocols for further investigations of patients according to the clinical presentation.

Clinical presentation of hyperandrogenism In a newborn female infant, clitoromegaly is a sign of androgen excess in utero. In some cases, virilisation is so profound that without careful inspection, the child can get male gender assignment. Ambiguous genitalia can also reflect a failing of 46XY male development, so karyotyping is important. Androgen excess in a child and adult can cause changes in body hair, growth patterns and apocrine sweat gland function. Bone age may be advanced by excess androgen exposure in children although before the end of puberty the normal growth spurt is aborted by epiphyseal fusion and the adult patient will be short. Adrenarche needs to be considered. This is a period of development characterised by the appearance of pubic and axillary hair and increase in body odour in girls around 8 years of age and boys around 9 years of age.7 Early adrenarche (premature adrenarche/pubarche) is a condition reflecting early development of the zona reticularis in the adrenal gland between the cortex and medulla secreting androgen.8 CAH and adrenal tumours need to be excluded. Imaging will be needed in some patients. Premature adrenarche, once characterised, is usually a benign diagnosis. The hair growth pattern in adrenarche in the pubic and axillary areas is different to the general hair growth (hypertrichosis) with use of certain drugs

such as cyclosporin, diazoxide, minoxidil and phenytoin. The hair pattern is also different to the androgenic pattern seen in adrenal tumours, polycystic ovary syndrome (PCOS) and other states where hair is on the arms, legs, back and chin. Puberty results when pulsatile secretion of gonadotrophins activates the gonads. In precocious puberty, the secondary sex characteristics develop prematurely. This can include breast development in girls (oestrogen exposure) and growth of penis, facial hair and musculature in boys from androgen excess without the normal pubertal changes in ovaries seen on ultrasound and size of the testes. Hirsutism is defined as excess body or coarse facial hair in females in a male-like pattern. Hair growth should be assessed on chin, face, chest, abdomen, back, thighs and upper arms. A scoring system by Ferriman and Gallwey in 19619 is a visual assessment and a recent version with patient photographs is a useful reference point.10 A polycystic ovarian appearance is inevitable in any of these clinical situations.11,12 PCOS is also often associated with insulin resistance and hyperinsulinemia. Insulin resistance may also develop through the metabolic effects of androgens.7,13,14 The precise mechanism for cardiovascular risk is still unclear. In the adolescent and adult, androgen excess may be due to peripheral or central defects and tend to be rarer in origin. Anovulation, menstrual irregularity and infertility in CAH are also linked with the androgen and progesterone excess.15 The hypothalamic-pituitary ovarian axis is disrupted and gonadotrophin releasing hormone (GnRH) pulse frequency is rapid, favouring LH secretion. Disorders of dihydrotestosterone (DHT) production may also manifest for the first time at 12–15 years of age through symptoms of androgen excess. In rare cases, there may be additional dysmorphic features. Exogenous steroid use needs to be questioned. A dialogue between clinician and the laboratory is important in the diagnosis of an ever broadening field. A multidisciplinary team meeting should assemble clinical and laboratory experts to debate issues around difficult cases.16

Steroid synthesis Adrenal steroid synthesis of cortisol and aldosterone from cholesterol (Figure 1) starts with the uptake of cholesterol into mitochondria through the action of steroidogenic acute regulatory protein (StAR) followed by side chain cleavage (a cytochrome P-450 enzyme, now called CYP11A1), to liberate pregnenolone.17 Cholesterol and pregnenolone have -hydroxyl groups at carbon 3 (C3) and double bonds at C5 to C6 (5-ene). In the smooth endoplasmic reticulum of the adrenal zona fasciculata, pregnenolone can be hydroxylated at

Downloaded from acb.sagepub.com at UNIV TORONTO on July 2, 2014

Figure 1. Biosynthesis of steroids, including alternative path to DHT from 17-OHP via androsterone and 5a-androstane-3a,17-diol.

426 Annals of Clinical Biochemistry 51(4)

Downloaded from acb.sagepub.com at UNIV TORONTO on July 2, 2014

Honour

427

C17 by 17a-hydroxylase (CYP17A1) to produce 17-hydroxypregnenolone which is converted to the steroid hormone 17-OHP (17-hydroxy 4-pregnene-3,20dione) with carbonyl at C-3 and C4 to C5 double bond (4-ene) by the action of 3-hydroxysteroid dehydrogenase and 5,4-isomerase (HSD3B2). 17-OHP is converted to 11 deoxycortisol by C-21 hydroxylase (CYP21A2). 11-deoxycortisol has to migrate to the mitochondria for 11-hydroxylation by 11 beta hydroxylase (CYP11B1) to produce cortisol in the fasciculata zone. 17-OHP is thus an intermediate steroid in the adrenal pathway from cholesterol to cortisol but not significantly for androgens because 17-OHP is a poor substrate for CYP17A1. Cortisol and cortisone are interconverted by oxidase and reductase activities of two 11-hydroxysteroid dehydrogenases (HSD11B2 and B1 respectively). The electron transfer of the reductase is coupled with hexose-6-phosphate dehydrogenase.18 In the outer zona glomerulosa, pregnenolone is converted to progesterone, deoxycorticosterone, corticosterone and aldosterone through the actions of HSD3B2, CYP21A2, and then CYP11B2 that has 18-hydroxylase and aldosterone synthetase activity. In the human adrenal cortex, CYP17A1 can efficiently convert pregnenolone to dehydroepiandrosterone (DHA) but cannot convert progesterone to androstenedione. DHA is conjugated with sulphuric acid through the action of sulphotransferase to the sulphate (DHAS). In adults, DHAS is the main steroid secreted by the adrenal cortex from the inner zona reticularis leading to circulating concentrations of 2–10 micromoles per litre. The adrenals secrete small amounts of testosterone but greater amounts of androstenedione through the action of CYP11A1, HSD3B2 and 17-hydroxysteroid dehydrogenase (an aldo-keto reductase, AKR1C3).19 The gonads also produce 17OHP and high concentrations of 17-OHP can be found for example in ovarian follicles.20 17-OHP may also be converted to DHT through a ‘backdoor path’.21,22 This may provide a more efficient route to DHT under conditions of high 17-OHP accumulation as in CAH due to 21-hydroxylase deficiency.23 In the past decade, other aspects of steroid synthesis have attracted clinical interest. A cytochrome P450-oxidoreductase (POR) is part of the electron transport system for CYP21A2 and CYP17 (and other enzymes).24

Congenital adrenal hyperplasia Increased concentrations of the 17-OHP in the peripheral circulation indicate a deficiency of 21-hydroxylase activity.25 Failure to synthesise cortisol leads to an increase in ACTH through activation of the cortisol negative feedback and is the basis of CAH. Aldosterone synthesis can also be affected, so

progesterone may also be elevated failing its conversion to deoxycorticosterone. There are variable consequences to glucose and electrolyte problems due to steroid deficiencies that need to be characterised. Electrolyte disturbance is the key feature of the male neonate with CAH. The classical form of 21-hydroxylase deficiency presents usually in childhood. Virilisation is the result of hyperandrogenaemia, and in many cases salt loss is the consequence of reduced aldosterone synthesis. The androgen excess in CAH must derive from 17-hydroxypregnenolone to produce DHA through the 17,20-lyase activity of CYP17A1. The non-classical CAH form (NC-CAH) was first called late-onset (sometimes cryptic) because of the time of presentation notably in adolescents and adults. The effects of cortisol and aldosterone deficiency are not usually apparent and the presentation is due to signs of androgen excess. Precocious puberty, acne, hirsutism and subfertility are the commonest presenting features of NC-CAH requiring investigations. The prevalence of NC-CAH in women with androgen excess can be from 0.6 to 9% according to ethnic background and genotype (see reference 26 for survey). The disorder is more common in Ashkenazi Jews, Mediterranean, Middle-Eastern and Indian populations. NC-CAH is more likely to be suspected clinically in females.

Genetic basis of 21-hydroxylase deficiency The gene for steroid 21-hydroxylase was sequenced in 1985,27–29 some 6 years before genetic mutations specific to mild forms of 21-hydroxylase deficiency were described in family studies that included asymptomatic members.30–31 The clinical presentation are usually related to the magnitude of 21-hydroxylase deficiency, being severe in neonates and milder during childhood and adults. Two CYP21 genes are located in chromosome 6 within the coding region for the major histocompatability complex and fourth component of complement, C4. The active 21-hydroxylase gene (CYP21A2) is about 30 kb downstream of a pseudogene (CYP21A1P) that does not encode protein. The genes have 98% nucleotide homology in 10 exons and 9% homology in introns.27 Human leucocyte antigen (HLA) typing has been used to track gene mutations of CYP21A2. HLA-B14 is found in about 40% of NCCAH haplotypes. The common mutations of CYP21A2 are genetic recombinations with CYP21A1P as well as deletions and point mutations. The mutations cause varying degrees of enzyme deficit as shown by in vitro expression studies32 as well as genotype–phenotype correlations.33–34 In the classic defect, more than 95% of enzyme activity is lost through mutations in the CYP21A2 gene coding the

Downloaded from acb.sagepub.com at UNIV TORONTO on July 2, 2014

428

Annals of Clinical Biochemistry 51(4)

protein.27 Where the enzyme activity is lost, the phenotype is of salt-wasting disease with virilisation. When 21-hydroxylase activity is as little as 2% of normal, the phenotype is simple virilisation. If the 21-hydroxylase activity is 10–75% of normal, patients have the mild NC-CAH picture. This was recently reviewed.35 Work with molecular models of the enzyme substituted for the absence of crystalline structure of the human 21-hydroxylase protein.36–39 NC-CAH is associated most commonly with homozygous Valine to Leucine conversion at position 281 that alters haem binding. This mutation is found in NC-CAH often as a compound heterozygote with a severe mutation on the other allele. Another mutation commonly found in NC-CAH is Proline to Leucine at position 30 that is at the point for membrane insertion and enzyme stability. The NC-CAH mutations have been found to affect 21-hydroxylase activity at oxidoreductase interactions, salt-bridge and hydrogen bonding networks and non-conserved hydrophobic interactions.40 Multiple copies of the genes are found due to unequal crossover in meiosis at various points in the sequence.41 The approach for genetic analysis of gene deletions, gene conversions, point mutations and small gene alterations has been reviewed recently.42 A reference laboratory skilled in this analysis should be consulted. Different concentrations of service are offered from linkage analysis to complete sequencing of the gene. Analyses are available for detection of deletions, duplications and targetted mutations. The impact of changes in the gene on protein expression may be required in a few cases. CAH is now recognised as a spectrum of clinical presentations from mild androgen excess (NCCAH) to prominent androgen excess with adrenal failure of cortisol (simple virilising form) and often of aldosterone (salt-wasting form) production.

Related enzyme defects Androgen excess can also be the consequence of defects of HSD3B, CYP11B1 and POR. Cases are usually detected after birth because of typical presentations. The markers for these defects are DHAS, 11-deoxycortisol and in the case of POR both 17-OHP and corticosterone. In CAH due to 3-hydroxysteroid dehydrogenase (HSD3B2) deficiency, the aldosterone production may also be blocked and, in the absence of this mineralocorticoid, salt loss leads to stimulation of renin production.43 DHA is the substrate for HSD3B2 and a circulating marker for the defect along with 17-hydroxypregnenolone. In a newborn infant with CAH due to HSD3B2, the 17-OHP can be apparently elevated either through cross reaction in an immunoassay with 17-hydroxypregnenolone (and more so it’s sulphate form) or because 17-OHP

is genuinely raised through the action of other 3-hydroxysteroid dehydrogenase isoenzymes on 17hydroxypregnenolone.44 In CAH due to 11-hydroxylase deficiency, the production of deoxycorticosterone is increased and this potent mineralocorticoid causes renal sodium retention with suppression of renin release form the renal juxtaglomerular apparatus.45 ‘Functional’ deficiency of 21-hydroxylase and 17hydroxylase has been described with POR defects.46,47 17-OHP and corticosterone concentrations in serum are raised although assays for the latter are not generally available. Although POR deficiency is usually detected in neonates with ambiguous genitalia and skeletal abnormalities (Antley-Bixler syndrome), mild forms can present with adrenal insufficiency and later polycystic ovaries. A urinary steroid profile (USP) shows characteristic high excretion of both 17-OHP and corticosterone metabolites.47 Non-classic forms of CAH may also be attributable to mutations leading to mild defects in HSD3B248 and CYP11B149–51, POR52 and StAR.53 The interconversion of cortisol to cortisone can be defective in the reverse reaction by cortisone reductase (HSD11B1) and leads to a need for an increase in ACTH to raise cortisol and androgen production. This condition presents like adrenarche but is best characterised in a USP from the high ratio of cortisone to cortisol metabolites.54 This can be distinguished from apparent cortisone reductase deficiency (ACRD) due to inactivating mutations in hexose-6-phosphate dehydrogenase gene by even lower cortisol metabolites and higher cortisone metabolites. High ACTH also leads to some increased 17-OHP production.

17-OHP analysis The first assay for 17-OHP per se was based on competitive protein binding with radioactive 17-OHP using a crude preparation of cortisol binding globulin as ligand.55,56 Radioimmunoassays were available from 197157 and have remained the prominent test although are now being superceded by liquid chromatography coupled with tandem mass spectrometry (LC-MS/MS) methods.58,59 Two main analytical methods are thus in use for plasma 17-OHP; immunoassay and chromatographic. Immunoassay (IA) is the most frequently used technique for steroid measurement. 17-OHP is quantitatively displaced from its binding proteins and measured immunometrically using antibodies supposedly specific to 17-OHP. In practice, some crossreactivity, e.g. with 11-deoxycortisol, is inevitable. Commercial assays that use antibody-coated tubes are designed for 17-OHP concentrations up to 100 nmol/L; reflecting the more common requirement, in clinical

Downloaded from acb.sagepub.com at UNIV TORONTO on July 2, 2014

Honour

429

Table 1. 17-OHP results with RIA and GC-MS (Supraregional Assay Service data) basally and after ACTH stimulation.

Infants (unstressed) Infants (stressed) Adult males Adut females – follicular phase Adult females – luteal phase Patients with untreated 21-hydroxylase deficiency Patients with NC- CAH Heterozygote carriers

RIA (serum) nmol/L (60 min ACTH stim

GC-MS (plasma)nmol/L (30 min ACTH stim)

100 (63–470) (6–44)

30 nmol/L indicates non-classic CAH (depending on the assay and reference data). A number of other steroid defects in steroid synthesis and metabolism can be characterised by a USP with, in some cases, further genetic tests for absolute confirmation. All laboratory results should be interpreted using reference ranges for the appropriate assay technology. This is particularly important when steroid assays of greater specificity (for example, LC-MS/MS) are introduced to the laboratory repertoire.

1. Bongiovanni AM and Root AW. The adrenogenital syndrome. NEJM 1963; 268: 1283–1289. 2. Butler GC and Marrian GF. The isolation of pregnane3,17,20-triol from the urine of women showing the adrenogenital syndrome. J Biol Chem 1937; 119: 565–570. 3. Hechter O and Pincus G. The genesis of the adrenocortical secretion. Physiol Rev 1954; 34: 459–496. 4. Lieberman S. The generally accepted version of steroidogenesis is not free of uncertainties: other tenable and possibly superior renditions may be invented. J Steroid Biochem Mol Biol 2008; 109: 197–199. 5. Eberlein WR and Bongiovanni AM. Partial characterization of urinary adrenocortical steroids in adrenal hyperplasia. J Clin Invest 1955; 34: 1337–1343. 6. Stanczyk FZ. Diagnosis of hyperandrogenism: biochemical criteria. Best Pract Res Clin Endocrinol Metab 2006; 20: 177–191. 7. Williams RM, Ward CE and Hughes IA. Premature adrenarche. Arch Dis Child 2012; 97: 250–254. 8. Honour JW, Kelnar CJ and Brook CG. Urine steroid excretion rates in childhood reflect growth and activity of the adrenal cortex. Acta Endocrinol (Copenh) 1991; 124: 219–224. 9. Ferriman D and Gallwey JD. Clinical assessment of body hair growth in women. J Clin Endocrinol Metab 1961; 21: 1440–1447. 10. Yildiz BO, Bolour S, Woods K, et al. Visually scoring hirsutism. Hum Reprod Update 2010; 16: 51–64. 11. Hassa H, Tanir HM and Yildiz Z. Comparison of clinical and laboratory characteristics of cases with polycystic ovarian syndrome based on Rotterdam’s criteria and women whose only clinical signs are oligo/anovulation or hirsutism. Arch Gynecol Obstet 2006; 274: 227–232. 12. Pall M, Azziz R, Beires J, et al. The phenotype of hirsute women: a comparison of polycystic ovary syndrome and 21-hydroxylase-deficient nonclassic adrenal hyperplasia. Fertil Steril 2010; 94: 684–689. 13. Schnaider-Rezek GS, Lemos-Marini SH, Baptista MT, et al. Metabolic evaluation of young women with congenital adrenal hyperplasia. Arq Bras Endocrinol Metabol 2011; 55: 646–652. 14. Han TS, Krone N, Willis DS, et al. United Kingdom Congenital adrenal Hyperplasia Adult StudyExecutive (CaHASE). Quality of life in adults with congenital adrenal hyperplasiarelates to glucocorticoid treatment, adiposity and insulin resistance: United Kingdom Congenital adrenal Hyperplasia Adult Study Executive (CaHASE). Eur J Endocrinol 2013; 168: 887–893. 15. Holmes-Walker DJ, Conway GS, Honour JW, et al. Menstrual disturbance and hypersecretion of progesterone in women with congenital adrenal hyperplasia due to 21-hydroxylase deficiency. Clin Endocrinol (Oxf) 1995; 43: 291–296.

Acknowledgements The author is grateful to members of the Clinical Science Review Committee, Dr Gill Rumsby and Dr Adel A Ismail for advice and constructive criticism. This article was prepared at the invitation of the Clinical Sciences Reviews Committee of the Association for Clinical Biochemistry and Laboratory Medicine.

Declaration of conflicting interests None.

Funding This research received no specific grant from any funding agency in the public, commercial, or not-for-profit sectors.

Ethical approval Not applicable.

Guarantor JWH.

JWH researched the literature and is responsible for the manuscript in full.

Downloaded from acb.sagepub.com at UNIV TORONTO on July 2, 2014

436

Annals of Clinical Biochemistry 51(4)

16. Brain CE, Creighton SM, Mushtaq I, et al. Holistic management of DSD. Best Pract Res Clin Endocrinol Metab 2010; 24: 335–354. 17. Ghayee HK and Auchus RJ. Basic concepts and recent developments in human steroid hormone biosynthesis. Rev Endocr Metab Disord 2007; 8: 290–300. 18. Draper N, Walker EA, Bujalska IJ, et al. Mutations in the genes encoding 11beta-hydroxysteroid dehydrogenase type 1 and hexose-6-phosphate dehydrogenase interact to cause cortisone reductase deficiency. Nat Genet 2003; 34: 434–439. 19. Nakamura Y, Hornsby PJ, Casson P, et al. Type 5 17beta-hydroxysteroid dehydrogenase (AKR1C3) contributes to testosterone production in the adrenal reticularis. J Clin Endocrinol Metab 2009; 94: 2192–2198. 20. Amin M, Simerman A, Cho M, et al. 21-Hydroxylasederived steroids in follicles of nonobese women undergoing ovarian stimulation for in vitro fertilization (IVF) positively correlate with lipid content of luteinized granulosa cells (LGCs) as a source of cholesterol for steroid synthesis. J Clin Endocrinol Metab 2014; Jan 13:jc20133204. [Epub ahead of print]. 21. Auchus RJ. Non-traditional metabolic pathways of adrenal steroids. Rev Endocr Metab Disord 2009; 10: 27–32. 22. Penning TM. New frontiers in androgen biosynthesis and metabolism. Curr Opin Endocrinol Diabet Obes 2010; 17: 233–239. 23. Kamrath C, Hartmann MF and Wudy SA. Androgen synthesis in patients with congenital adrenal hyperplasia due to 21-hydroxylase deficiency. Horm Metab Res 2013; 45: 86–91. 24. Miller WL. Minireview: regulation of steroidogenesis by electron transfer. Endocrinology 2005; 146: 2544–2550. 25. Barnes ND and Atherden SM. Diagnosis of congenital adrenal hyperplasia by measurement of plasma 17-hydroxyprogesterone. Arch Dis Child 1972; 47: 62–65. 26. Witchel SF and Azziz R. Nonclassic congenital adrenal hyperplasia. Int J Pediatr Endocrinol 2010; 2010: 625105. 27. White PC, New MI and Dupont B. Molecular cloning of steroid 21-hydroxylase. Ann N Y Acad Sci 1985; 458: 277–287. 28. Higashi Y, Yoshioka H, Yamane M, et al. Complete nucleotide sequence of two steroid 21-hydroxylase genes tandemly arranged in human chromosome: a pseudogene and a genuine gene. Proc Natl Acad Sci U S A 1986; 83: 2841–2845. 29. White PC, New MI and Dupont B. Structure of human steroid 21-hydroxylase genes. Proc Natl Acad Sci U S A 1986; 83: 5111–5115. 30. Tusie-Luna MT, Speiser PW, Dumic M, et al. A mutation (Pro-30 to Leu) in CYP21 represents a potential nonclassic steroid 21-hydroxylase deficiency allele. Mol Endocrinol 1991; 5: 685–692. 31. Helmberg A, Tusie-Luna MT, Tabarelli M, et al. R339H and P453S: CYP21 mutations associated with nonclassic steroid 21-hydroxylase deficiency that are not apparent gene conversions. Mol Endocrinol 1992; 6: 1318–1322. 32. Tusie-Luna MT, Traktman P and White PC. Determination of functional effects of mutations in the

33.

34.

35. 36.

37.

38.

39.

40.

41.

42.

43.

44.

45.

46.

47.

48.

steroid 21-hydroxylase gene (CYP21) using recombinant vaccinia virus. J Biol Chem 1990; 265: 20916–20922. Rumsby G, Avey CJ, Conway GS, et al. Genotype-phenotype analysis in late onset 21-hydroxylase deficiency in comparison to the classical forms. Clin Endocrinol (Oxf) 1998; 48: 707–711. New MI, Abraham M, Gonzalez B, et al. Genotype-phenotype correlation in 1,507 families with congenital adrenal hyperplasia owing to 21-hydroxylase deficiency. Proc Natl Acad Sci U S A 2013; 110: 2611–2616. Witchel SF and Azziz R. Nonclassic congenital adrenal hyperplasia. Int J Pediatr Endocrinol 2010; 2010: 625105. Mornet E and Gibrat JF. A 3D model of human P450c21: study of the putative effects of steroid 21-hydroxylase gene mutations. Hum Genet 2000; 106: 330–339. Bojunga J, Welsch C, Antes I, et al. Structural and functional analysis of a novel mutation of CYP21B in a heterozygote carrier of 21-hydroxylase deficiency. Hum Genet 2005; 117: 558–564. Robins T, Carlsson J, Sunnerhagen M, et al. Molecular model of human CYP21 based on mammalian CYP2C5: structural features correlate with clinical severity of mutations causing congenital adrenal hyperplasia. Mol Endocrinol 2006; 20: 2946–2964. Zhao B, Lei L, Kagawa N, et al. Three-dimensional structure of steroid 21-hydroxylase (cytochrome P450 21A2) with two substrates reveals locations of diseaseassociated variants. J Biol Chem 2012; 287: 10613–10622. Haider S, Islam B, D’Atri V, et al. Structure-phenotype correlations of human CYP21A2 mutations in congenital adrenal hyperplasia. Proc Natl Acad Sci U S A 2013; 110: 2605–2610. Werkmeister JW, New MI, Dupont B, et al. Frequent deletion and duplication of the steroid 21-hydroxylase genes. Am J Hum Genet 1986; 39: 461–469. Krone N and Arlt W. Genetics of congenital adrenal hyperplasia. Best Pract Res Clin Endocrinol Metab 2009; 23: 181–192. Bongiovanni AM and Kellenbenz G. The adrenogenital syndrome with deficiency of 3-hydroxystroid dehydrogenase. J Clin Invest 41: 2086–2092. Sanchez R, Rhe´aume E, Laflamme N, et al. Detection and functional characterization of the novel missense mutation Y254D in type II 3 beta-hydroxysteroid dehydrogenase (3 beta HSD) gene of a female patient with nonsalt-losing 3 beta HSD deficiency. J Clin Endocrinol Metab 1994; 78: 561–567. Nimkarn S and New MI. Steroid 11beta-hydroxylase deficiency congenital adrenal hyperplasia. Trens Endocrinol Metab 2008; 19: 96–99. Flu¨ck CE and Miller WL. P450 oxidoreductase deficiency: a new form of congenital adrenal hyperplasia. Curr Opin Pediatr 2006; 18: 435–441. Krone N, Reisch N and Idkowiak J. Genotypephenotype analysis in congenital adrenal hyperplasia due to P450 oxidoreductase deficiency. J Clin Endocrinol Metab 2012; 97: E257–E267. Lutfallah C, Wang W, Mason JI, et al. Newly proposed hormonal criteria via genotypic proof for type II

Downloaded from acb.sagepub.com at UNIV TORONTO on July 2, 2014

Honour

49.

50.

51.

52.

53.

54.

55.

56.

57.

58.

59.

60.

61.

62.

63.

437

3beta-hydroxysteroid dehydrogenase deficiency. J Clin Endocrinol Metab 2002; 87: 2611–2622. Joehrer K, Geley S, Strasser-Wozak EM, et al. CYP11B1 mutations causing non-classic adrenal hyperplasia due to 11 beta-hydroxylase deficiency. Hum Mol Genet 1997; 6: 1829–1834. Parajes S, Loidi L, Reisch N, et al. Functional consequences of seven novel mutations in the CYP11B1 gene: four mutations associated with nonclassic and three mutations causing classic 11{beta}-hydroxylase deficiency. J Clin Endocrinol Metab 2010; 95: 779–788. Reisch N, Ho¨gler W, Parajes S, et al. A diagnosis not to be missed: Non-classic steroid 11-hydroxylase deficiency presenting with premature adrenarche and hirsutism. J Clin Endocrinol Metab 2013; [Epub ahead of print]. Sahakitrungruang T, Huang N, Tee MK, et al. Clinical, genetic, and enzymatic characterization of P450 oxidoreductase deficiency in four patients. J Clin Endocrinol Metab 2009; 94: 4992–5000. Metherell LA, Naville D, Halaby G, et al. Nonclassic lipoid congenital adrenal hyperplasia masquerading as familial glucocorticoid deficiency. J Clin Endocrinol Metab 2009; 94: 3865–3871. Lavery GG, Idkowiak J, Sherlock M, et al. Novel H6PDH mutations in two girls with premature adrenarche: ‘apparent’ and ‘true’ CRD can be differentiated by urinary steroid profiling. Eur J Endocrinol 2013; 168: K19–K26. Strott CA and Lipsett MB. Measurement of 17-hydroxyprogesterone. J Clin Endocrinol Metab 1968; 28: 1426–1430. Strott CA, Yoshimi T, Bardin CW, et al. Blood progesterone and 17-hydroxyprogesterone leves and production rates in a boy with virilizing congenital adrenal hyperplasia. J Clin Endocrinol Metab 1968; 28: 1085–1088. Abraham GE, Swerdloff RS, Tulchinsky D, et al. Radioimmunoassay of plasma 17-hydroxyprogesterone. J Clin Endocrinol Metab 1971; 33: 42–46. Wudy SA, Hartmann M and Svoboda M. Determination of 17-hydroxyprogesterone in plasma by stable isotope dilution/benchtop liquid chromatography-tandem mass spectrometry. Horm Res 2000; 53: 68–71. Turpeinen U, Itkonen O, Ahola L, et al. Determination of 17alpha-hydroxyprogesterone in serum by liquid chromatography-tandem mass spectrometry and immunoassay. Scand J Clin Lab Invest 2005; 65: 3–12. Wong T, Shackleton CH, Covey TR, et al. Identification of the steroids in neonatal plasma that interfere with 17 alpha-hydroxyprogesterone radioimmunoassays. Clin Chem 1992; 38: 1830–1837. Boudi A, Giton F, Galons H, et al. Development of a plasma 17alpha-hydroxyprogesterone time resolved fluorescence immunoassay involving a biotinylated tracer. Steroids 2000; 65: 103–108. Tripathi V, Naran S, Chaube SK, et al. Development of rapid and sensitive one-step direct enzyme immunosorbent assay for 17-alpha-OH-progesterone in serum. J Immunoassay Immunochem 2008; 29: 117–127. Chong H, Cheah SH, Ragavan M, et al. Development of an indirect enzyme immunoassay using monoclonal

64.

65.

66.

67.

68.

69.

70.

71.

72.

73.

74.

75.

76.

antibodies for the measurement of 17alpha-hydroxyprogesterone in human serum. J Immunoassay Immunochem 2009; 30: 166–179. Wei JQ, Zhou XT and Wei JI. Simultaneous measurement of eight corticosteroids by liquid chromatography, and application of the procedure to diagnosis of congenital adrenal hyperplasia. Clin Chem 1987; 33: 1354–1359. Courant F, Aksglaede L, Antignac JP, et al. Assessment of circulating sex steroid levels in prepubertal and pubertal boys and girls by a novel ultrasensitive gas chromatography-tandem mass spectrometry method. J Clin Endocrinol Metab 2010; 95: 82–92. Masse´ R and Wright LA. Proposed definitive methods for measurement of plasma testosterone and 17 alphahydroxyprogesterone. Clin Biochem 1996; 29: 321–331. Honour JW. Development and validation of a quantitative assay based on tandem mass spectrometry. Ann Clin Biochem 2011; 48: 97–111. Wudy SA, Wachter UA, Homoki J, et al. 17 alphahydroxyprogesterone, 4-androstenedione, and testosterone profiled by routine stable isotope dilution/gas chromatography-mass spectrometry in plasma of children. Pediatr Res 1995; 38: 76–80. Guo T, Chan M and Soldin SJ. Steroid profiles using liquid chromatography-tandem mass spectrometry with atmospheric pressure photoionization source. Arch Pathol Lab Med 2004; 128: 469–475. Rauh M, Gro¨schl M, Rascher W, et al. Automated, fast and sensitive quantification of 17 alpha-hydroxyprogesterone, androstenedione and testosterone by tandem mass spectrometry with on-line extraction. Steroids 2006; 71: 450–458. Garagorri JM, Rodrı´ guez G, Lario-Elboj AJ, et al. Reference levels for 17-hydroxyprogesterone, 11-desoxycortisol, cortisol, testosterone, dehydroepiandrosterone sulfate and androstenedione in infants from birth to six months of age. Eur J Pediatr 2008; 167: 647–653. Magnisali P, Dracopoulou M, Mataragas M, et al. Routine method for the simultaneous quantification of 17alpha-hydroxyprogesterone, testosterone, dehydroepiandrosterone, androstenedione, cortisol, and pregnenolone in human serum of neonates using gas chromatography–mass spectrometry. J Chromatogr A 2008; 1206: 166–177. Ceglarek U, Kortz L, Leichtle A, et al. Rapid quantification of steroid patterns in human serum by on-line solid phase extraction combined with liquid chromatographytriple quadrupole linear ion trap mass spectrometry. Clin Chim Acta 2009; 401: 114–118. Rossi C, Calton L, Hammond G, et al. Serum steroid profiling for congenital adrenal hyperplasia using liquid chromatography-tandem mass spectrometry. Clin Chim Acta 2010; 411: 222–228. Dhillon K, Ho T, Rich P, et al. An automated method on analysis of blood steroids using liquid chromatography tandem mass spectrometry: application to population screening for congenital adrenal hyperplasia in newborns. Clin Chim Acta 2011; 412: 2076–2084. Rossi C, Calton L, Brown HA, et al. Confirmation of congenital adrenal hyperplasia by adrenal steroid

Downloaded from acb.sagepub.com at UNIV TORONTO on July 2, 2014

438

77.

78.

79.

80.

81.

82.

83.

84.

85.

86.

87.

88.

Annals of Clinical Biochemistry 51(4) profiling of filter paper dried blood samples using ultraperformance liquid chromatography-tandem mass spectrometry. Clin Chem Lab Med 2011; 49: 677–684. Koal T, Schmiederer D, Pham-Tuan H, et al. Standardized LC-MS/MS based steroid hormone profile-analysis. J Steroid Biochem Mol Biol 2012; 129: 129–138. Soldin OP, Sharma H, Husted L, et al. Pediatric reference intervals for aldosterone, 17alpha-hydroxyprogesterone, dehydroepiandrosterone, testosterone and 25-hydroxy vitamin D3 using tandem mass spectrometry. Clin Biochem 2009; 42: 823–827. Barth JH, Field HP, Yasmin E, et al. Defining hyperandrogenism in polycystic ovary syndrome: measurement of testosterone and androstenedione by liquid chromatography-tandem mass spectrometry and analysis by receiver operator characteristic plots. Eur J Endocrinol 2010; 162: 611–615. Kulle AE, Riepe FG, Melchior D, et al. A novel ultrapressure liquid chromatography tandem mass spectrometry method for the simultaneous determination of androstenedione, testosterone, and dihydrotestosterone in pediatric blood samples: age- and sex-specific reference data. J Clin Endocrinol Metab 2010; 95: 2399–2409. Kushnir MM, Blamires T, Rockwood AL, et al. Liquid chromatography-tandem mass spectrometry assay for androstenedione, dehydroepiandrosterone, and testosterone with pediatric and adult reference intervals. Clin Chem 2010; 56: 1138–1147. Rothman MS, Carlson NE, Xu M, et al. Reexamination of testosterone, dihydrotestosterone, estradiol and estrone levels across the menstrual cycle and in postmenopausal women measured by liquid chromatographytandem mass spectrometry. Steroids 2011; 76: 177–182. Fanelli F, Belluomo I, Di Lallo VD, et al. Serum steroid profiling by isotopic dilution-liquid chromatography– mass spectrometry: comparison with current immunoassays and reference intervals in healthy adults. Steroids 2011; 76: 244–253. Janse F, Eijkemans MJ, Goverde AJ, et al. Assessment of androgen concentration in women: liquid chromatography-tandem mass spectrometry and extraction RIA show comparable results. Eur J Endocrinol 2011; 165: 925–933. Haring R, Hannemann A, John U, et al. Age-specific reference ranges for serum testosterone and androstenedione concentrations in women measured by liquid chromatography-tandem mass spectrometry. J Clin Endocrinol Metab 2012; 97: 408–415. Rhea JM, French D and Molinaro RJ. Direct total and free testosterone measurement by liquid chromatography tandem mass spectrometry across two different platforms. Clin Biochem 2013; 46: 656–664. Kyriakopoulou L, Yazdanpanah M, Colantonio DA, et al. A sensitive and rapid mass spectrometric method for the simultaneous measurement of eight steroid hormones and CALIPER pediatric reference intervals. Clin Biochem 2013; 46: 642–651. Kulle AE, Welzel M, Holterhus PM, et al. Implementation of a liquid chromatography tandem mass spectrometry assay for eight adrenal C-21 steroids

89.

90.

91.

92.

93.

94.

95.

96.

97.

98.

99.

100.

and pediatric reference data. Horm Res Paediatr 2013; 79: 22–31. Broccardo CJ, Schauer KL, Kohrt WM, et al. Multiplexed analysis of steroid hormones in human serum using novel microflow tile technology and LCMS/MS. J Chromatogr B Analyt Technol Biomed Life Sci 2013; 934: 16–21. Fanelli F, Gambineri A, Belluomo I, et al. Androgen profiling by liquid chromatography-tandem mass spectrometry (LC-MS/MS) in healthy normal-weight ovulatory and anovulatory late adolescent and young women. J Clin Endocrinol Metab 2013; 98: 3058–3067. Meier U, Schnabel C, Kunz D, et al. Comparison of three commercial assays for the measurement of 17alpha-hydroxyprogesterone (17alpha-OHPR): limitations of the quality control system. Clin Chem Lab Med 2004; 42: 450–454. Etter ML, Eichorst J and Lehotay DC. Clinical determination of 17-hydroxyprogesterone in serum by LCMS/MS comparison to Coat-A-Count RIA method. J Chromatogr B Analyt Technol Biomed Sci 2006; 840: 69–74. New MI. Extensive clinical experience: nonclassical 21-hydroxylase deficiency. J Clin Endocrinol Metab 2006; 91: 4205–4214. Erratum in: J Clin Endocrinol Metab 2007; 92: 142. Dosage error in article text. Lashansky G, Saenger P, Fishman K, et al. Normative data for adrenal steroidogenesis in a healthy pediatric population: age- and sex-related changes after adrenocorticotropin stimulation. J Clin Endocrinol Metab 1991; 73: 674–686. Azziz R, Hincapie LA, Knochenhauer ES, et al. Screening for 21-hydroxylase-deficient nonclassic adrenal hyperplasia among hyperandrogenic women: a prospective study. Fertil Steril 1999; 72: 915–925. Hague WM, Honour JW, Adams J, et al. Steroid responses to ACTH in women with polycystic ovaries. Clin Endocrinol (Oxf) 1989; 30: 355–365. Bridges NA, Hindmarsh PC, Pringle PJ, et al. Cortisol, androstenedione (A4), dehydroepiandrosterone sulphate (DHAS) and 17-hydroxyprogesterone (17OHP) responses to low doses of (1-24)ACTH. J Clin Endocrinol Metab 1998; 83: 3750–3753. Erratum in: J Clin Endocrinol Metab 1999; 84: 2972. J Clin Endocrinol Metab 1999; 84: 4177. Rosenfield RL, Barnes RB and Ehrmann DA. Studies of the nature of 17-hydroxyprogesterone hyperresponsiveness to gonadotropin-releasing hormone agonist challenge in functional ovarian hyperandrogenism. J Clin Endocrinol Metab 1994; 79: 1686–1692. Laven JS, Imani B, Eijkemans MJ, et al. New approach to polycystic ovary syndrome and other forms of anovulatory infertility. Obstet Gynecol Surv 2002; 57: 755–767. Fiet J, Villette JM, Galons H, et al. The application of a new highly-sensitive radioimmunoassay for plasma 21-deoxycortisol to the detection of steroid-21hydroxylase deficiency. Ann Clin Biochem 1994; 31: 56–64.

Downloaded from acb.sagepub.com at UNIV TORONTO on July 2, 2014

Honour

439

101. Lee MM, Misra M, Donahoe PK, et al. MIS/AMH in the assessment of cryptorchidism and intersex conditions. Mol Cell Endocrinol 2003; 211: 91–98. 102. Jones RL, Owen LJ, Adaway JE, et al. Simultaneous analysis of cortisol and cortisone in saliva using XLCMS/MS for fully automated online solid phase extraction. J Chromatogr B Analyt Technol Biomed Life Sci 2012; 881–882: 42–48. 103. Krone N, Hughes BA, Lavery GG, et al. Gas chromatography/mass spectrometry (GC/MS) remains a preeminent discovery tool in clinical steroid investigations even in the era of fast liquid chromatographytandem mass spectrometry (LC/MS/MS). J Steroid Biochem Mol Biol 2010; 121: 496–504. 104. Dhillon K, Ho T, Rich P, et al. An automated method on analysis of blood steroids using liquid chromatography tandem mass spectrometry: application to population screening for congenital adrenal hyperplasia in newborns. Clin Chim Acta 2011; 412: 2076–2084. 105. Janzen N, Sander S, Terhardt M, et al. Fast and direct quantification of adrenal steroids by tandem mass spectrometry in serum and dried blood spots. J Chromatogr B Analyt Technol Biomed Life Sci 2008; 861: 117–122. 106. Janzen N, Sander S, Terhardt M, et al. Rapid steroid hormone quantification for congenital adrenal hyperplasia (CAH) in dried blood spots using UPLC liquid chromatography-tandem mass spectrometry. Steroids 2011; 76: 1437–1442. 107. Magnisali P, Chalioti MB, Livadara T, et al. Simultaneous quantification of 17a-OH progesterone, 11-deoxycortisol, 4-androstenedione, cortisol and cortisone in newborn blood spots using liquid chromatography-tandem mass spectrometry. J Chromatogr B Analyt Technol Biomed Life Sci 2011; 879: 1565–1572. 108. Seo JY, Park HD, Kim JW, et al. Steroid profiling for congenital adrenal hyperplasia by tandem mass spectrometry as a second-tier test reduces follow-up burdens in a tertiary care hospital: A retrospective and prospective evaluation. J Perinat Med 2013; Aug 29: 1-7. DOI: 10.1515/jpm-2013-0154. [Epub ahead of print]. 109. Cavarzere P, Samara-Boustani D, Flechtner I, et al. Transient hyper-17-hydroxyprogesteronemia: a clinical subgroup of patients diagnosed at neonatal screening for congenital adrenal hyperplasia. Eur J Endocrinol 2009; 161: 285–292. 110. Christakoudi S, Cowan DA, Christakudis G, et al. 21Hydroxylase deficiency in the neonate – trends in steroid anabolism and catabolism during the first weeks of life. J Steroid Biochem Mol Biol 2013; 138C: 334–347. 111. Koyama Y, Homma K, Miwa M, et al. Measurement of reference intervals for urinary free adrenal steroid levels in Japanese newborn infants by using stable isotope dilution gas chromatography/mass spectrometry. Clin Chim Acta 2013; 415: 302–305. 112. Christakoudi S, Cowan DA and Taylor NF. A new marker for early diagnosis of 21-hydroxylase deficiency:3beta, 16alpha, 17alpha-trihydroxy-5alpha-

113.

114.

115.

116. 117.

118.

119.

120.

121.

122.

123.

124. 125.

pregnane-7,20-dione. J Steroid Biochem Mol Biol 2010; 121: 574–581. Koyama Y, Homma K, Fukami M, et al. Two-step biochemical differential diagnosis of classic 21-hydroxylase deficiency and cytochrome P450 oxidoreductase deficiency in Japanese infants by GC-MS measurement of urinary pregnanetriolone/tetrahydrocortisone ratio and 11-hydroxyandrosterone. Clin Chem 2012; 58: 741–747. Ratcliffe WA, McClure JP, Auld WH, et al. Precocious pseudopuberty due to a rare form of congenital adrenal hyperplasia. Biochemical investigation and pitfalls in interpretation of hormone assays. Ann Clin Biochem 1982; 19: 145–150. Flint JL and Jacobson JD. Adrenal hypoplasia congenita presenting as congenital adrenal hyperplasia. Case Rep Endocrinol 2013; 2013: 393584. Honour JW, Price DA and Grant DB. Virilizing adrenocortical tumors in childhood. Pediatrics 1986; 78: 547. Wolthers OD, Cameron FJ, Scheimberg I, et al. Androgen secreting adrenocortical tumours. Arch Dis Child 1999; 80: 46–50. Knowles RL, Khalid JM, Oerton JM, et al. Late clinical presentation of congenital adrenal hyperplasia in older children: findings from national paediatric surveillance. Arch Dis Child 2013; Sep 16. DOI:10.1136/archdischild2012-303070. [Epub ahead of print] PubMed PMID: 24043550. Dunger DB, Villa AK, Matthews DR, et al. Pattern of secretion of bioactive and immunoreactive gonadotrophins in normal pubertal children. Clin Endocrinol (Oxf) 1991; 35: 267–275. Bevan BR, Savvas M, Jenkins JM, et al. Abnormal steroid excretion in gestational trophoblastic disease complicated by ovarian theca-lutein cysts. J Clin Pathol 1986; 39: 627–634. Rosenfield RL, Mortensen M, Wroblewski K, et al. Determination of the source of androgen excess in functionally atypical polycystic ovary syndrome by a short dexamethasone androgen-suppression test and a lowdose ACTH test. Hum Reprod 2011; 26: 3138–3146. Fukami M, Shozu M, Soneda S, et al. Aromatase excess syndrome: identification of cryptic duplications and deletions leading to gain of function of CYP19A1 and assessment of phenotypic determinants. J Clin Endocrinol Metab 2011; 96: E1035–E1043. Wajchenberg BL, Achando SS, Okada H, et al. Determination of the source(s) of androgen overproduction in hirsutism associated with polycystic ovary syndrome by simultaneous adrenal and ovarian venous catheterization. Comparison with the dexamethasone suppression test. J Clin Endocrinol Metab 1986; 63: 1204–1210. Arora MK, Yadav A and Saini V. Role of hormones in acne vulgaris. Clin Biochem 2011; 44: 1035–1040. Ostlere LS, Rumsby G, Holownia P, et al. Carrier status for steroid 21-hydroxylase deficiency is only one factor

Downloaded from acb.sagepub.com at UNIV TORONTO on July 2, 2014

440

126.

127.

128.

129.

130.

131.

132.

133.

134.

Annals of Clinical Biochemistry 51(4) in the variable phenotype of acne. Clin Endocrinol (Oxf) 1998; 48: 209–215. Maimoun L, Philibert P, Cammas B, et al. Phenotypical, biological, and molecular heterogeneity of 5a-reductase deficiency: an extensive international experience of 55 patients. J Clin Endocrinol Metab 2011; 96: 296–307. Bertelloni S, Balsamo A, Giordani L, et al. 17betaHydroxysteroid dehydrogenase-3 deficiency: from pregnancy to adolescence. J Endocrinol Invest 2009; 32: 666–670. Kulshreshtha B, Philibert P, Eunice M, et al. Phenotype, hormonal profile and genotype of subjects with partial androgen insensitivity syndrome: report of a family with four adult males and one child with disorder of sexual differentiation. Andrologia 2009; 41: 257–263. Hugues JN, Theron-Gerard L, Coussieu C, et al. Assessment of theca cell function prior to controlled ovarian stimulation: the predictive value of serum basal/stimulated steroid levels. Hum Reprod 2010; 25: 228–234. Camats N, Pandey AV, Ferna´ndez–Cancio M, et al. STAR splicing mutations cause the severe phenotype of lipoid congenital adrenal hyperplasia: insights from a novel splice mutation and review of reported cases. Clin Endocrinol (Oxf) 2013; Jul 16. DOI:10.1111/ cen.12293. [Epub ahead of print] PubMed PMID: 23859637. Arlt W, Biehl M, Taylor AE, et al. Urine steroid metabolomics as a biomarker tool for detecting malignancy in adrenal tumors. J Clin Endocrinol Metab 2011; 96: 3775–3784. Auchus RJ and Arlt W. Approach to the patient: the adult with congenital adrenal hyperplasia. J Clin Endocrinol Metab 2013; 98: 2645–2655. Trapp CM and Oberfield SE. Recommendations for treatment of nonclassic congenital adrenal hyperplasia (NCCAH): an update. Steroids 2012; 77: 342–346. Matthews D and Cheetham T. What is the best approach to the teenage patient presenting with nonclassical congenital adrenal hyperplasia: should we always treat with glucocorticoids? Clin Endocrinol (Oxf) 2013; 78: 338–341.

135. Kamrath C, Bo¨hles H and Maser-Gluth C. Comparison of two different radioimmunoassays to measure 17-hydroxyprogesterone during treatment monitoring of children with congenital adrenal hyperplasia due to 21-hydroxylase deficiency. Clin Chim Acta 2011; 412: 186–189. 136. Tripathi V, Nara S, Singh K, et al. A competitive immunochromatographic strip assay for 17-a-hydroxy progesterone using colloidal gold nanoparticles. Clin Chim Acta 2012; 413: 262–268. 137. Whitaker MJ, Debono M, Huatan H, et al. An oral multiparticulate, modified-release, hydrocortisone replacement therapy that provides physiological cortisol exposure. Clin Endocrinol (Oxf) 2013; Aug 27. DOI: 10.1111/cen.12316. [Epub ahead of print] PubMed PMID: 23980724. 138. Bryan SM, Honour JW and Hindmarsh PC. Management of altered hydrocortisone pharmacokinetics in a boy with congenital adrenal hyperplasia using a continuous subcutaneous hydrocortisone infusion. J Clin Endocrinol Metab 2009; 94: 3477–3480. 139. Azziz R, Carmina E, Dewailly D, et al. Task force on the phenotype of the polycystic ovary syndrome of the Androgen Excess and PCOS Society. The Androgen Excess and PCOS Society criteria for the polycystic ovary syndrome: the complete task force report. Fertil Steril 2009; 91: 456–488. 140. Speiser PW, Azziz R, Baskin LS, et al Congenital adrenal hyperplasia due to steroid 21-hydroxylase deficiency: an Endocrine Society clinical practice guideline. J Clin Endocrinol Metab 2010; 95: 4133–4160. Erratum in: J Clin Endocrinol Metab 2010; 95: 5137. 141. Rotterdam ESHRE/ASRM-Sponsored PCOS consensus workshop group. Revised 2003 consensus on diagnostic criteria and long-term health risks related to polycystic ovary syndrome (PCOS). Hum Reprod 2004; 19: 41–47. 142. Dunaif A and Fauser B. Renaming PCOS – a two state solution. J Clin Endocrinol Metab 2013; Sep 5. [Epub ahead of print] PubMed PMID: 4009134.

Downloaded from acb.sagepub.com at UNIV TORONTO on July 2, 2014

17-Hydroxyprogesterone in children, adolescents and adults.

17-Hydroxyprogesterone (17-OHP) is an intermediate steroid in the adrenal biosynthetic pathway from cholesterol to cortisol and is the substrate for s...
456KB Sizes 53 Downloads 3 Views