Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Peter C. Hindmarsh is active.

Publication


Featured researches published by Peter C. Hindmarsh.


Clinical Endocrinology | 2003

Blood pressure in children and adolescents with congenital adrenal hyperplasia (21-hydroxylase deficiency): a preliminary report.

Edna Roche; Evangelia Charmandari; Mehul T. Dattani; Peter C. Hindmarsh

background Classical congenital adrenal hyperplasia (CAH) is characterized by a defect in cortisol and aldosterone secretion, adrenal hyperandrogenism, impaired adrenal medullary function and insulin insensitivity. The latter along with the increased tendency towards obesity raises questions whether other cardiovascular risk factors are altered in CAH.


Best Practice & Research Clinical Endocrinology & Metabolism | 2009

Management of the child with congenital adrenal hyperplasia

Peter C. Hindmarsh

Classical congenital adrenal hyperplasia (CAH) due to 21-hydroxylase deficiency leads to glucocorticoid and mineralocorticoid deficiency. Management should be viewed as a process of care which requires input from an interdisciplinary team. Glucocorticoid therapy should take the form of hydrocortisone in a starting dose of 15 mg/m(2)/day (divided into three doses), and the dose should be titrated to blood or urine profiles of cortisol and 17-hydroxyprogesterone. Mineralocorticoid replacement (9 alpha-fludrocortisone) requires higher doses in infancy and childhood than in adolescence. The starting dose should be 150 microg/m(2)/day, and the dose thereafter titrated to plasma renin activity and blood pressure. Despite adequate glucocorticoid substitution and concordance with medical therapy, control can be difficult during puberty due to alterations in the clearance of hydrocortisone, and dosing schedules may need to be adjusted to account for this. Follow-up should address the many facets of CAH, which should be assessed at an annual review, and a suggested protocol is presented.


The Journal of Clinical Endocrinology and Metabolism | 2012

Genotype-phenotype analysis in congenital adrenal hyperplasia due to P450 oxidoreductase deficiency

Nils Krone; Nicole Reisch; Jan Idkowiak; Vivek Dhir; Hannah E Ivison; Beverly Hughes; Ian T. Rose; Donna M. O'Neil; Raymon Vijzelaar; Matthew J. Smith; Fiona MacDonald; Trevor R. Cole; Nicolai Adolphs; John S. Barton; Edward Blair; Stephen R. Braddock; Felicity Collins; Deborah L. Cragun; Mehul T. Dattani; Ruth Day; Shelley Dougan; Miriam Feist; Michael Gottschalk; John Welbourn Gregory; Michaela Haim; Rachel Harrison; Anne Haskins Olney; Berthold P. Hauffa; Peter C. Hindmarsh; Robert J. Hopkin

Context: P450 oxidoreductase deficiency (PORD) is a unique congenital adrenal hyperplasia variant that manifests with glucocorticoid deficiency, disordered sex development (DSD), and skeletal malformations. No comprehensive data on genotype-phenotype correlations in Caucasian patients are available. Objective: The objective of the study was to establish genotype-phenotype correlations in a large PORD cohort. Design: The design of the study was the clinical, biochemical, and genetic assessment including multiplex ligation-dependent probe amplification (MLPA) in 30 PORD patients from 11 countries. Results: We identified 23 P450 oxidoreductase (POR) mutations (14 novel) including an exonic deletion and a partial duplication detected by MLPA. Only 22% of unrelated patients carried homozygous POR mutations. p.A287P was the most common mutation (43% of unrelated alleles); no other hot spot was identified. Urinary steroid profiling showed characteristic PORD metabolomes with variable impairment of 17α-hydroxylase and 21-hydroxylase. Short cosyntropin testing revealed adrenal insufficiency in 89%. DSD was present in 15 of 18 46,XX and seven of 12 46,XY individuals. Homozygosity for p.A287P was invariably associated with 46,XX DSD but normal genitalia in 46,XY individuals. The majority of patients with mild to moderate skeletal malformations, assessed by a novel scoring system, were compound heterozygous for missense mutations, whereas nearly all patients with severe malformations carried a major loss-of-function defect on one of the affected alleles. Conclusions: We report clinical, biochemical, and genetic findings in a large PORD cohort and show that MLPA is a useful addition to POR mutation analysis. Homozygosity for the most frequent mutation in Caucasians, p.A287P, allows for prediction of genital phenotype and moderate malformations. Adrenal insufficiency is frequent, easily overlooked, but readily detected by cosyntropin testing.


Clinical Endocrinology | 2007

Severe loss-of-function mutations in the adrenocorticotropin receptor (ACTHR, MC2R) can be found in patients diagnosed with salt-losing adrenal hypoplasia

Lin Lin; Peter C. Hindmarsh; Louise A. Metherell; Mahmoud Alzyoud; Maryam Alali; Caroline Brain; Adrian J. L. Clark; Mehul T. Dattani; John C. Achermann

Objective  Familial glucocorticoid deficiency type I (FGD1) is a rare form of primary adrenal insufficiency resulting from recessive mutations in the ACTH receptor (MC2R, MC2R). Individuals with this condition typically present in infancy or childhood with signs and symptoms of cortisol insufficiency, but disturbances in the renin‐angiotensin system, aldosterone synthesis or sodium homeostasis are not a well‐documented association of FGD1. As ACTH stimulation has been shown to stimulate aldosterone release in normal controls, and other causes of hyponatraemia can occur in children with cortisol deficiency, we investigated whether MC2R changes might be identified in children with primary adrenal failure who were being treated for mineralocorticoid insufficiency.


The Journal of Clinical Endocrinology and Metabolism | 2009

Management of Altered Hydrocortisone Pharmacokinetics in a Boy with Congenital Adrenal Hyperplasia Using a Continuous Subcutaneous Hydrocortisone Infusion

Sinead Bryan; John W. Honour; Peter C. Hindmarsh

BACKGROUND Conventional hydrocortisone dosing schedules do not mimic the normal circadian rhythm of cortisol, making it difficult to optimize treatment in congenital adrenal hyperplasia (CAH). CASE DETAILS We report a 14.5-year-old boy with CAH who had reduced bioavailability [42% (normal 80% orally and 100% by im route)] and increased clearance [half-life 50 min (normal range, 70-100 min)] of oral doses of hydrocortisone leading to ambient serum 17-hydroxyprogesterone concentrations of 400 nmol/liter (14.5 ng/ml) and androstenedione concentrations of 24.9 nmol/liter (7.1 ng/ml). INTERVENTION Using a continuous but variable sc hydrocortisone infusion via an insulin pump, rapid control of his CAH was attained with a normal cortisol circadian profile. Average daily hydrocortisone dose was 17.4-18.6 mg/m(2), which produces on average 24-h serum cortisol and 17-hydroxyprogesterone concentrations of 316 nmol/liter (115 ng/ml) and 4.3 nmol/liter (1.4 ng/ml), respectively. Therapy has been maintained over 4 yr with suppression of normal adrenal androgen production and normal progression through puberty. CONCLUSIONS Continuous sc infusion of hydrocortisone may prove a valuable adjunct to therapy for CAH, particularly in patients requiring high doses of oral hydrocortisone and in those with abnormal hydrocortisone pharmacokinetics.


BMJ | 2011

Effect of oxandrolone and timing of pubertal induction on final height in Turner’s syndrome: randomised, double blind, placebo controlled trial

Emma Jane Gault; Rebecca J Perry; T. J. Cole; Sarah Casey; Wendy F. Paterson; Peter C. Hindmarsh; Peter Betts; David B. Dunger; Malcolm Donaldson

Objective To examine the effect of oxandrolone and the timing of pubertal induction on final height in girls with Turner’s syndrome receiving a standard dose of growth hormone. Design Randomised, double blind, placebo controlled trial. Setting 36 paediatric endocrinology departments in UK hospitals. Participants Girls with Turner’s syndrome aged 7-13 years at recruitment, receiving recombinant growth hormone therapy (10 mg/m2/week). Interventions Participants were randomised to oxandrolone (0.05 mg/kg/day, maximum 2.5 mg/day) or placebo from 9 years of age. Those with evidence of ovarian failure at 12 years were further randomised to oral ethinylestradiol (year 1, 2 µg daily; year 2, 4 μg daily; year 3, 4 months each of 6, 8, and 10 μg daily) or placebo; participants who received placebo and those recruited after the age of 12.25 years started ethinylestradiol at age 14. Main outcome measure Final height. Results 106 participants were recruited, of whom 14 withdrew and 82/92 reached final height. Both oxandrolone and late pubertal induction increased final height: by 4.6 (95% confidence interval 1.9 to 7.2) cm (P=0.001, n=82) for oxandrolone and 3.8 (0.0 to 7.5) cm (P=0.05, n=48) for late pubertal induction with ethinylestradiol. In the 48 children who were randomised twice, the effects on final height (compared with placebo and early induction of puberty) of oxandrolone alone, late induction alone, and oxandrolone plus late induction were similar, averaging 7.1 (3.4 to 10.8) cm (P<0.001). No cases of virilisation were reported. Conclusion Oxandrolone had a positive effect on final height in girls with Turner’s syndrome treated with growth hormone, as did late pubertal induction with ethinylestradiol at age 14 years. However, these effects were not additive, so using both had no advantage. Oxandrolone could, therefore, be offered as an alternative to late pubertal induction for increasing final height in Turner’s syndrome. Trial registration Current Controlled Trials ISRCTN50343149.


Movement Disorders | 2014

Plasma melatonin is reduced in Huntington's disease

Eirini Kalliolia; Edina Silajdžić; Rajasree Nambron; Nathan R. Hill; Anisha Doshi; Chris Frost; Hilary Watt; Peter C. Hindmarsh; Maria Björkqvist; Thomas T. Warner

This study was undertaken to determine whether the production of melatonin, a hormone regulating sleep in relation to the light/dark cycle, is altered in Huntingtons disease. We analyzed the circadian rhythm of melatonin in a 24‐hour study of cohorts of control, premanifest, and stage II/III Huntingtons disease subjects. The mean and acrophase melatonin concentrations were significantly reduced in stage II/III Huntingtons disease subjects compared with controls. We also observed a nonsignificant trend toward reduced mean and acrophase melatonin in premanifest Huntingtons disease subjects. Onset of melatonin rise was significantly more temporally spread in both premanifest and stage II/III Huntingtons disease subjects compared with controls. A nonsignificant trend also was seen for reduced pulsatile secretion of melatonin. Melatonin concentrations are reduced in Huntingtons disease. Altered melatonin patterns may provide an explanation for disrupted sleep and circadian behavior in Huntingtons disease, and represent a biomarker for disease state. Melatonin therapy may help the sleep disorders seen in Huntingtons disease.


Archives of Disease in Childhood | 2001

Congenital adrenal hyperplasia: management during critical illness

Evangelia Charmandari; E J Lichtarowicz-Krynska; Peter C. Hindmarsh; Atholl Johnston; A Aynsley-Green; C. G. D. Brook

BACKGROUND Little is known of the optimal dose and administration schedule of hydrocortisone in critically ill patients with congenital adrenal hyperplasia (CAH) caused by 21-hydroxylase deficiency. AIM To determine plasma cortisol concentrations after intravenous administration of hydrocortisone in children with CAH and to relate these to plasma cortisol concentrations achieved by endogenous secretion in the stress of critical illness in previously healthy children. METHODS Plasma cortisol concentrations were measured in 20 patients with classical CAH (median age 11.2 years, range 6.1–16.4) following intravenous administration of hydrocortisone 15 mg/m2; and in 60 critically ill mechanically ventilated children (median age 2.5 years, range 0.25–16.3) on admission to the paediatric intensive care unit and for 24 hours thereafter. RESULTS In the CAH patients, plasma cortisol reached a mean peak of 1648.3 nmol/l (SD 511.9) within 10 minutes of the intravenous bolus, and fell rapidly thereafter; levels remained greater than 450 nmol/l for 2.5 hours only. In critically ill children, mean plasma cortisol on admission to the intensive care unit was 727 nmol/l (SD 426.1). Cortisol concentrations remained raised during the first 24 hours. CONCLUSIONS Critically ill patients with classical CAH may be best managed with a single intravenous hydrocortisone bolus followed by a constant rate infusion of hydrocortisone.


Clinical Endocrinology | 2014

Cardiovascular risk factors in children and adolescents with congenital adrenal hyperplasia due to 21-hydroxylase deficiency

Anbezhil Subbarayan; Mehul T. Dattani; Catherine Peters; Peter C. Hindmarsh

The prevalence of cardiovascular risk factors in congenital adrenal hyperplasia (CAH) varies widely. In the light of recent changes in treatment regimens, we have reassessed the prevalence of these risk factors in our current cohort of patients with CAH due to P450c21 deficiency.


Clinical Endocrinology | 2015

The relationship between Insulin-like Growth Factor 1, sex steroids and timing of the pubertal growth spurt

T. J. Cole; Ml Ahmed; Ma Preece; Peter C. Hindmarsh; David B. Dunger

Progress through puberty involves a complex hormonal cascade, but the individual contributions of hormones, particularly IGF‐1, are unknown. We reanalysed Chard growth study data to explore the tempo of puberty based on changes in both height and hormone levels, using a novel method of growth curve analysis.

Collaboration


Dive into the Peter C. Hindmarsh's collaboration.

Top Co-Authors

Avatar

Evangelia Charmandari

National and Kapodistrian University of Athens

View shared research outputs
Top Co-Authors

Avatar

Mehul T. Dattani

UCL Institute of Child Health

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Carol Dezateux

University College London

View shared research outputs
Top Co-Authors

Avatar

Juliet Oerton

UCL Institute of Child Health

View shared research outputs
Top Co-Authors

Avatar

Rachel L Knowles

UCL Institute of Child Health

View shared research outputs
Top Co-Authors

Avatar

Atholl Johnston

Queen Mary University of London

View shared research outputs
Top Co-Authors

Avatar

Eirini Kalliolia

UCL Institute of Neurology

View shared research outputs
Top Co-Authors

Avatar

Rajasree Nambron

UCL Institute of Neurology

View shared research outputs
Top Co-Authors

Avatar

Thomas T. Warner

UCL Institute of Neurology

View shared research outputs
Researchain Logo
Decentralizing Knowledge