1
|
Balagamage C, Arshad A, Elhassan YS, Ben Said W, Krone RE, Gleeson H, Idkowiak J. Management aspects of congenital adrenal hyperplasia during adolescence and transition to adult care. Clin Endocrinol (Oxf) 2024; 101:332-345. [PMID: 37964596 DOI: 10.1111/cen.14992] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Revised: 10/11/2023] [Accepted: 11/03/2023] [Indexed: 11/16/2023]
Abstract
The adolescent period is characterised by fundamental hormonal changes, which affect sex steroid production, cortisol metabolism and insulin sensitivity. These physiological changes have a significant impact on patients with congenital adrenal hyperplasia (CAH). An essential treatment aim across the lifespan in patients with CAH is to replace glucocorticoids sufficiently to avoid excess adrenal androgen production but equally to avoid cardiometabolic risks associated with excess glucocorticoid intake. The changes to the hormonal milieu at puberty, combined with poor adherence to medical therapy, often result in unsatisfactory control exacerbating androgen excess and increasing the risk of metabolic complications due to steroid over-replacement. With the physical and cognitive maturation of the adolescent with CAH, fertility issues and sexual function become a new focus of patient care in the paediatric clinic. This requires close surveillance for gonadal dysfunction, such as irregular periods/hirsutism or genital surgery-associated symptoms in girls and central hypogonadism or testicular adrenal rest tumours in boys. To ensure good health outcomes across the lifespan, the transition process from paediatric to adult care of patients with CAH must be planned carefully and early from the beginning of adolescence, spanning over many years into young adulthood. Its key aims are to empower the young person through education with full disclosure of their medical history, to ensure appropriate follow-up with experienced physicians and facilitate access to multispecialist teams addressing the complex needs of patients with CAH.
Collapse
Affiliation(s)
- Chamila Balagamage
- Centre for Endocrinology, Diabetes and Metabolism, Birmingham Health Partners, University of Birmingham, Birmingham, UK
- Department of Endocrinology and Diabetes, Birmingham Children's Hospital, Birmingham Women's and Children's NHS Foundation Trust, Birmingham, UK
| | - Amynta Arshad
- Centre for Endocrinology, Diabetes and Metabolism, Birmingham Health Partners, University of Birmingham, Birmingham, UK
- The Medical School, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Yasir S Elhassan
- Centre for Endocrinology, Diabetes and Metabolism, Birmingham Health Partners, University of Birmingham, Birmingham, UK
- Department of Endocrinology, University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
- Institute of Metabolism and Systems Research, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Wogud Ben Said
- Centre for Endocrinology, Diabetes and Metabolism, Birmingham Health Partners, University of Birmingham, Birmingham, UK
- Department of Endocrinology and Diabetes, Birmingham Children's Hospital, Birmingham Women's and Children's NHS Foundation Trust, Birmingham, UK
- Institute of Metabolism and Systems Research, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Ruth E Krone
- Centre for Endocrinology, Diabetes and Metabolism, Birmingham Health Partners, University of Birmingham, Birmingham, UK
- Department of Endocrinology and Diabetes, Birmingham Children's Hospital, Birmingham Women's and Children's NHS Foundation Trust, Birmingham, UK
| | - Helena Gleeson
- Centre for Endocrinology, Diabetes and Metabolism, Birmingham Health Partners, University of Birmingham, Birmingham, UK
- Department of Endocrinology, University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
| | - Jan Idkowiak
- Centre for Endocrinology, Diabetes and Metabolism, Birmingham Health Partners, University of Birmingham, Birmingham, UK
- Department of Endocrinology and Diabetes, Birmingham Children's Hospital, Birmingham Women's and Children's NHS Foundation Trust, Birmingham, UK
- Institute of Metabolism and Systems Research, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| |
Collapse
|
2
|
Bacila IA, Lawrence NR, Badrinath SG, Balagamage C, Krone NP. Biomarkers in congenital adrenal hyperplasia. Clin Endocrinol (Oxf) 2024; 101:300-310. [PMID: 37608608 DOI: 10.1111/cen.14960] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Revised: 07/25/2023] [Accepted: 08/07/2023] [Indexed: 08/24/2023]
Abstract
Monitoring of hormone replacement therapy represents a major challenge in the management of congenital adrenal hyperplasia (CAH). In the absence of clear guidance and standardised monitoring strategies, there is no consensus among clinicians regarding the relevance of various biochemical markers used in practice, leading to wide variability in their application and interpretation. In this review, we summarise the published evidence on biochemical monitoring of CAH. We discuss temporal variations of the most commonly measured biomarkers throughout the day, the interrelationship between different biomarkers, as well as their relationship with different glucocorticoid and mineralocorticoid treatment regimens and clinical outcomes. Our review highlights significant heterogeneity across studies in both aims and methodology. However, we identified key messages for the management of patients with CAH. The approach to hormone replacement therapy should be individualised, based on the individual hormonal profile throughout the day in relation to medication. There are limitations to using 17-hydroxyprogesterone, androstenedione and testosterone, and the role of additional biomarkers such 11-oxygenated androgens which are more disease specific should be further established. Noninvasive monitoring via salivary and urinary steroid measurements is becoming increasingly available and should be considered, especially in the management of children with CAH. Additionally, this review indicates the need for large scale longitudinal studies analysing the interrelation between different monitoring strategies used in clinical practice and health outcomes in children and adults with CAH.
Collapse
Affiliation(s)
| | - Neil R Lawrence
- Department of Oncology and Metabolism, University of Sheffield, Sheffield, UK
| | | | - Chamila Balagamage
- Department of Endocrinology, Birmingham Women's & Children's Hospital, Birmingham, UK
- Department of Endocrinology, Sheffield Children's Hospital, Sheffield, UK
| | - Nils P Krone
- Department of Oncology and Metabolism, University of Sheffield, Sheffield, UK
- Department of Endocrinology, Sheffield Children's Hospital, Sheffield, UK
| |
Collapse
|
3
|
Sarafoglou K, Kim MS, Lodish M, Felner EI, Martinerie L, Nokoff NJ, Clemente M, Fechner PY, Vogiatzi MG, Speiser PW, Auchus RJ, Rosales GBG, Roberts E, Jeha GS, Farber RH, Chan JL. Phase 3 Trial of Crinecerfont in Pediatric Congenital Adrenal Hyperplasia. N Engl J Med 2024; 391:493-503. [PMID: 38828945 DOI: 10.1056/nejmoa2404655] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 06/05/2024]
Abstract
BACKGROUND Children with classic congenital adrenal hyperplasia (CAH) due to 21-hydroxylase deficiency require treatment with glucocorticoids, usually at supraphysiologic doses, to address cortisol insufficiency and reduce excess adrenal androgens. However, such treatment confers a predisposition to glucocorticoid-related complications. In 2-week phase 2 trials, patients with CAH who received crinecerfont, a new oral corticotropin-releasing factor type 1 receptor antagonist, had decreases in androstenedione levels. METHODS In this phase 3, multinational, randomized trial, we assigned pediatric participants with CAH, in a 2:1 ratio, to receive crinecerfont or placebo for 28 weeks. A stable glucocorticoid dose was maintained for 4 weeks, and the dose was then adjusted to a target of 8.0 to 10.0 mg per square meter of body-surface area per day (hydrocortisone dose equivalents), provided that the androstenedione level was controlled (≤120% of the baseline level or within the reference range). The primary efficacy end point was the change in the androstenedione level from baseline to week 4. A key secondary end point was the percent change in the glucocorticoid dose from baseline to week 28 while androstenedione control was maintained. RESULTS A total of 103 participants underwent randomization, of whom 69 were assigned to crinecerfont and 34 to placebo; 100 (97%) remained in the trial at 28 weeks. At baseline, the mean glucocorticoid dose was 16.4 mg per square meter per day, and the mean androstenedione level was 431 ng per deciliter (15.0 nmol/liter). At week 4, androstenedione was substantially reduced in the crinecerfont group (-197 ng per deciliter [-6.9 nmol/liter]) but increased in the placebo group (71 ng per deciliter [2.5 nmol/liter]) (least-squares mean difference [LSMD], -268 ng per deciliter [-9.3 nmol/liter]; P<0.001); the observed mean androstenedione value, obtained before the morning glucocorticoid dose, was 208 ng per deciliter (7.3 nmol/liter) in the crinecerfont group, as compared with 545 ng per deciliter (19.0 nmol/liter) in the placebo group. At week 28, the mean glucocorticoid dose had decreased (while androstenedione control was maintained) by 18.0% with crinecerfont but increased by 5.6% with placebo (LSMD, -23.5 percentage points; P<0.001). Headache, pyrexia, and vomiting were the most common adverse events. CONCLUSIONS In this phase 3 trial, crinecerfont was superior to placebo in reducing elevated androstenedione levels in pediatric participants with CAH and was also associated with a decrease in the glucocorticoid dose from supraphysiologic to physiologic levels while androstenedione control was maintained. (Funded by Neurocrine Biosciences; CAHtalyst Pediatric ClinicalTrials.gov number, NCT04806451.).
Collapse
Affiliation(s)
- Kyriakie Sarafoglou
- From the University of Minnesota Medical School and College of Pharmacy, Minneapolis (K.S.); Children's Hospital Los Angeles and Keck School of Medicine of USC, Los Angeles (M.S.K.), the University of California at San Francisco, Benioff Children's Hospital, San Francisco (M.L.), and Neurocrine Biosciences, San Diego (G.B.G.R., E.R., G.S.J., R.H.F., J.L.C.) - all in California; Emory University School of Medicine and Children's Healthcare of Atlanta, Atlanta (E.I.F.); Endocrinologie Pédiatrique, Centre de Référence Maladies Endocriniennes Rares de la Croissance et du Développement, Hôpital Universitaire Robert-Debré, Groupe Hospitalo-Universitaire de l'Assistance Publique-Hôpitaux de Paris Nord, and Université Paris Cité, Faculté de Santé, UFR de Médecine, Paris, and Université Paris-Saclay, INSERM Physiologie et Physiopathologie Endocriniennes, Le Kremlin-Bicêtre - all in France (L.M.); the University of Colorado School of Medicine, Children's Hospital Colorado, Aurora (N.J.N.); Pediatric Endocrinology, Hospital Universitario Vall d'Hebrón, Barcelona (M.C.); the University of Washington School of Medicine, Seattle Children's Hospital, Seattle (P.Y.F.); the Children's Hospital of Philadelphia, Philadelphia (M.G.V.); Cohen Children's Medical Center of NY, New Hyde Park, and the Donald and Barbara Zucker School of Medicine at Hofstra-Northwell, Hempstead - both in New York (P.W.S.); and the Departments of Pharmacology and Internal Medicine, Division of Metabolism, Endocrinology and Diabetes, University of Michigan Medical School, and the Endocrinology and Metabolism Section, Medicine Services, LTC Charles S. Kettles Veterans Affairs Medical Center - both in Ann Arbor (R.J.A.)
| | - Mimi S Kim
- From the University of Minnesota Medical School and College of Pharmacy, Minneapolis (K.S.); Children's Hospital Los Angeles and Keck School of Medicine of USC, Los Angeles (M.S.K.), the University of California at San Francisco, Benioff Children's Hospital, San Francisco (M.L.), and Neurocrine Biosciences, San Diego (G.B.G.R., E.R., G.S.J., R.H.F., J.L.C.) - all in California; Emory University School of Medicine and Children's Healthcare of Atlanta, Atlanta (E.I.F.); Endocrinologie Pédiatrique, Centre de Référence Maladies Endocriniennes Rares de la Croissance et du Développement, Hôpital Universitaire Robert-Debré, Groupe Hospitalo-Universitaire de l'Assistance Publique-Hôpitaux de Paris Nord, and Université Paris Cité, Faculté de Santé, UFR de Médecine, Paris, and Université Paris-Saclay, INSERM Physiologie et Physiopathologie Endocriniennes, Le Kremlin-Bicêtre - all in France (L.M.); the University of Colorado School of Medicine, Children's Hospital Colorado, Aurora (N.J.N.); Pediatric Endocrinology, Hospital Universitario Vall d'Hebrón, Barcelona (M.C.); the University of Washington School of Medicine, Seattle Children's Hospital, Seattle (P.Y.F.); the Children's Hospital of Philadelphia, Philadelphia (M.G.V.); Cohen Children's Medical Center of NY, New Hyde Park, and the Donald and Barbara Zucker School of Medicine at Hofstra-Northwell, Hempstead - both in New York (P.W.S.); and the Departments of Pharmacology and Internal Medicine, Division of Metabolism, Endocrinology and Diabetes, University of Michigan Medical School, and the Endocrinology and Metabolism Section, Medicine Services, LTC Charles S. Kettles Veterans Affairs Medical Center - both in Ann Arbor (R.J.A.)
| | - Maya Lodish
- From the University of Minnesota Medical School and College of Pharmacy, Minneapolis (K.S.); Children's Hospital Los Angeles and Keck School of Medicine of USC, Los Angeles (M.S.K.), the University of California at San Francisco, Benioff Children's Hospital, San Francisco (M.L.), and Neurocrine Biosciences, San Diego (G.B.G.R., E.R., G.S.J., R.H.F., J.L.C.) - all in California; Emory University School of Medicine and Children's Healthcare of Atlanta, Atlanta (E.I.F.); Endocrinologie Pédiatrique, Centre de Référence Maladies Endocriniennes Rares de la Croissance et du Développement, Hôpital Universitaire Robert-Debré, Groupe Hospitalo-Universitaire de l'Assistance Publique-Hôpitaux de Paris Nord, and Université Paris Cité, Faculté de Santé, UFR de Médecine, Paris, and Université Paris-Saclay, INSERM Physiologie et Physiopathologie Endocriniennes, Le Kremlin-Bicêtre - all in France (L.M.); the University of Colorado School of Medicine, Children's Hospital Colorado, Aurora (N.J.N.); Pediatric Endocrinology, Hospital Universitario Vall d'Hebrón, Barcelona (M.C.); the University of Washington School of Medicine, Seattle Children's Hospital, Seattle (P.Y.F.); the Children's Hospital of Philadelphia, Philadelphia (M.G.V.); Cohen Children's Medical Center of NY, New Hyde Park, and the Donald and Barbara Zucker School of Medicine at Hofstra-Northwell, Hempstead - both in New York (P.W.S.); and the Departments of Pharmacology and Internal Medicine, Division of Metabolism, Endocrinology and Diabetes, University of Michigan Medical School, and the Endocrinology and Metabolism Section, Medicine Services, LTC Charles S. Kettles Veterans Affairs Medical Center - both in Ann Arbor (R.J.A.)
| | - Eric I Felner
- From the University of Minnesota Medical School and College of Pharmacy, Minneapolis (K.S.); Children's Hospital Los Angeles and Keck School of Medicine of USC, Los Angeles (M.S.K.), the University of California at San Francisco, Benioff Children's Hospital, San Francisco (M.L.), and Neurocrine Biosciences, San Diego (G.B.G.R., E.R., G.S.J., R.H.F., J.L.C.) - all in California; Emory University School of Medicine and Children's Healthcare of Atlanta, Atlanta (E.I.F.); Endocrinologie Pédiatrique, Centre de Référence Maladies Endocriniennes Rares de la Croissance et du Développement, Hôpital Universitaire Robert-Debré, Groupe Hospitalo-Universitaire de l'Assistance Publique-Hôpitaux de Paris Nord, and Université Paris Cité, Faculté de Santé, UFR de Médecine, Paris, and Université Paris-Saclay, INSERM Physiologie et Physiopathologie Endocriniennes, Le Kremlin-Bicêtre - all in France (L.M.); the University of Colorado School of Medicine, Children's Hospital Colorado, Aurora (N.J.N.); Pediatric Endocrinology, Hospital Universitario Vall d'Hebrón, Barcelona (M.C.); the University of Washington School of Medicine, Seattle Children's Hospital, Seattle (P.Y.F.); the Children's Hospital of Philadelphia, Philadelphia (M.G.V.); Cohen Children's Medical Center of NY, New Hyde Park, and the Donald and Barbara Zucker School of Medicine at Hofstra-Northwell, Hempstead - both in New York (P.W.S.); and the Departments of Pharmacology and Internal Medicine, Division of Metabolism, Endocrinology and Diabetes, University of Michigan Medical School, and the Endocrinology and Metabolism Section, Medicine Services, LTC Charles S. Kettles Veterans Affairs Medical Center - both in Ann Arbor (R.J.A.)
| | - Laetitia Martinerie
- From the University of Minnesota Medical School and College of Pharmacy, Minneapolis (K.S.); Children's Hospital Los Angeles and Keck School of Medicine of USC, Los Angeles (M.S.K.), the University of California at San Francisco, Benioff Children's Hospital, San Francisco (M.L.), and Neurocrine Biosciences, San Diego (G.B.G.R., E.R., G.S.J., R.H.F., J.L.C.) - all in California; Emory University School of Medicine and Children's Healthcare of Atlanta, Atlanta (E.I.F.); Endocrinologie Pédiatrique, Centre de Référence Maladies Endocriniennes Rares de la Croissance et du Développement, Hôpital Universitaire Robert-Debré, Groupe Hospitalo-Universitaire de l'Assistance Publique-Hôpitaux de Paris Nord, and Université Paris Cité, Faculté de Santé, UFR de Médecine, Paris, and Université Paris-Saclay, INSERM Physiologie et Physiopathologie Endocriniennes, Le Kremlin-Bicêtre - all in France (L.M.); the University of Colorado School of Medicine, Children's Hospital Colorado, Aurora (N.J.N.); Pediatric Endocrinology, Hospital Universitario Vall d'Hebrón, Barcelona (M.C.); the University of Washington School of Medicine, Seattle Children's Hospital, Seattle (P.Y.F.); the Children's Hospital of Philadelphia, Philadelphia (M.G.V.); Cohen Children's Medical Center of NY, New Hyde Park, and the Donald and Barbara Zucker School of Medicine at Hofstra-Northwell, Hempstead - both in New York (P.W.S.); and the Departments of Pharmacology and Internal Medicine, Division of Metabolism, Endocrinology and Diabetes, University of Michigan Medical School, and the Endocrinology and Metabolism Section, Medicine Services, LTC Charles S. Kettles Veterans Affairs Medical Center - both in Ann Arbor (R.J.A.)
| | - Natalie J Nokoff
- From the University of Minnesota Medical School and College of Pharmacy, Minneapolis (K.S.); Children's Hospital Los Angeles and Keck School of Medicine of USC, Los Angeles (M.S.K.), the University of California at San Francisco, Benioff Children's Hospital, San Francisco (M.L.), and Neurocrine Biosciences, San Diego (G.B.G.R., E.R., G.S.J., R.H.F., J.L.C.) - all in California; Emory University School of Medicine and Children's Healthcare of Atlanta, Atlanta (E.I.F.); Endocrinologie Pédiatrique, Centre de Référence Maladies Endocriniennes Rares de la Croissance et du Développement, Hôpital Universitaire Robert-Debré, Groupe Hospitalo-Universitaire de l'Assistance Publique-Hôpitaux de Paris Nord, and Université Paris Cité, Faculté de Santé, UFR de Médecine, Paris, and Université Paris-Saclay, INSERM Physiologie et Physiopathologie Endocriniennes, Le Kremlin-Bicêtre - all in France (L.M.); the University of Colorado School of Medicine, Children's Hospital Colorado, Aurora (N.J.N.); Pediatric Endocrinology, Hospital Universitario Vall d'Hebrón, Barcelona (M.C.); the University of Washington School of Medicine, Seattle Children's Hospital, Seattle (P.Y.F.); the Children's Hospital of Philadelphia, Philadelphia (M.G.V.); Cohen Children's Medical Center of NY, New Hyde Park, and the Donald and Barbara Zucker School of Medicine at Hofstra-Northwell, Hempstead - both in New York (P.W.S.); and the Departments of Pharmacology and Internal Medicine, Division of Metabolism, Endocrinology and Diabetes, University of Michigan Medical School, and the Endocrinology and Metabolism Section, Medicine Services, LTC Charles S. Kettles Veterans Affairs Medical Center - both in Ann Arbor (R.J.A.)
| | - María Clemente
- From the University of Minnesota Medical School and College of Pharmacy, Minneapolis (K.S.); Children's Hospital Los Angeles and Keck School of Medicine of USC, Los Angeles (M.S.K.), the University of California at San Francisco, Benioff Children's Hospital, San Francisco (M.L.), and Neurocrine Biosciences, San Diego (G.B.G.R., E.R., G.S.J., R.H.F., J.L.C.) - all in California; Emory University School of Medicine and Children's Healthcare of Atlanta, Atlanta (E.I.F.); Endocrinologie Pédiatrique, Centre de Référence Maladies Endocriniennes Rares de la Croissance et du Développement, Hôpital Universitaire Robert-Debré, Groupe Hospitalo-Universitaire de l'Assistance Publique-Hôpitaux de Paris Nord, and Université Paris Cité, Faculté de Santé, UFR de Médecine, Paris, and Université Paris-Saclay, INSERM Physiologie et Physiopathologie Endocriniennes, Le Kremlin-Bicêtre - all in France (L.M.); the University of Colorado School of Medicine, Children's Hospital Colorado, Aurora (N.J.N.); Pediatric Endocrinology, Hospital Universitario Vall d'Hebrón, Barcelona (M.C.); the University of Washington School of Medicine, Seattle Children's Hospital, Seattle (P.Y.F.); the Children's Hospital of Philadelphia, Philadelphia (M.G.V.); Cohen Children's Medical Center of NY, New Hyde Park, and the Donald and Barbara Zucker School of Medicine at Hofstra-Northwell, Hempstead - both in New York (P.W.S.); and the Departments of Pharmacology and Internal Medicine, Division of Metabolism, Endocrinology and Diabetes, University of Michigan Medical School, and the Endocrinology and Metabolism Section, Medicine Services, LTC Charles S. Kettles Veterans Affairs Medical Center - both in Ann Arbor (R.J.A.)
| | - Patricia Y Fechner
- From the University of Minnesota Medical School and College of Pharmacy, Minneapolis (K.S.); Children's Hospital Los Angeles and Keck School of Medicine of USC, Los Angeles (M.S.K.), the University of California at San Francisco, Benioff Children's Hospital, San Francisco (M.L.), and Neurocrine Biosciences, San Diego (G.B.G.R., E.R., G.S.J., R.H.F., J.L.C.) - all in California; Emory University School of Medicine and Children's Healthcare of Atlanta, Atlanta (E.I.F.); Endocrinologie Pédiatrique, Centre de Référence Maladies Endocriniennes Rares de la Croissance et du Développement, Hôpital Universitaire Robert-Debré, Groupe Hospitalo-Universitaire de l'Assistance Publique-Hôpitaux de Paris Nord, and Université Paris Cité, Faculté de Santé, UFR de Médecine, Paris, and Université Paris-Saclay, INSERM Physiologie et Physiopathologie Endocriniennes, Le Kremlin-Bicêtre - all in France (L.M.); the University of Colorado School of Medicine, Children's Hospital Colorado, Aurora (N.J.N.); Pediatric Endocrinology, Hospital Universitario Vall d'Hebrón, Barcelona (M.C.); the University of Washington School of Medicine, Seattle Children's Hospital, Seattle (P.Y.F.); the Children's Hospital of Philadelphia, Philadelphia (M.G.V.); Cohen Children's Medical Center of NY, New Hyde Park, and the Donald and Barbara Zucker School of Medicine at Hofstra-Northwell, Hempstead - both in New York (P.W.S.); and the Departments of Pharmacology and Internal Medicine, Division of Metabolism, Endocrinology and Diabetes, University of Michigan Medical School, and the Endocrinology and Metabolism Section, Medicine Services, LTC Charles S. Kettles Veterans Affairs Medical Center - both in Ann Arbor (R.J.A.)
| | - Maria G Vogiatzi
- From the University of Minnesota Medical School and College of Pharmacy, Minneapolis (K.S.); Children's Hospital Los Angeles and Keck School of Medicine of USC, Los Angeles (M.S.K.), the University of California at San Francisco, Benioff Children's Hospital, San Francisco (M.L.), and Neurocrine Biosciences, San Diego (G.B.G.R., E.R., G.S.J., R.H.F., J.L.C.) - all in California; Emory University School of Medicine and Children's Healthcare of Atlanta, Atlanta (E.I.F.); Endocrinologie Pédiatrique, Centre de Référence Maladies Endocriniennes Rares de la Croissance et du Développement, Hôpital Universitaire Robert-Debré, Groupe Hospitalo-Universitaire de l'Assistance Publique-Hôpitaux de Paris Nord, and Université Paris Cité, Faculté de Santé, UFR de Médecine, Paris, and Université Paris-Saclay, INSERM Physiologie et Physiopathologie Endocriniennes, Le Kremlin-Bicêtre - all in France (L.M.); the University of Colorado School of Medicine, Children's Hospital Colorado, Aurora (N.J.N.); Pediatric Endocrinology, Hospital Universitario Vall d'Hebrón, Barcelona (M.C.); the University of Washington School of Medicine, Seattle Children's Hospital, Seattle (P.Y.F.); the Children's Hospital of Philadelphia, Philadelphia (M.G.V.); Cohen Children's Medical Center of NY, New Hyde Park, and the Donald and Barbara Zucker School of Medicine at Hofstra-Northwell, Hempstead - both in New York (P.W.S.); and the Departments of Pharmacology and Internal Medicine, Division of Metabolism, Endocrinology and Diabetes, University of Michigan Medical School, and the Endocrinology and Metabolism Section, Medicine Services, LTC Charles S. Kettles Veterans Affairs Medical Center - both in Ann Arbor (R.J.A.)
| | - Phyllis W Speiser
- From the University of Minnesota Medical School and College of Pharmacy, Minneapolis (K.S.); Children's Hospital Los Angeles and Keck School of Medicine of USC, Los Angeles (M.S.K.), the University of California at San Francisco, Benioff Children's Hospital, San Francisco (M.L.), and Neurocrine Biosciences, San Diego (G.B.G.R., E.R., G.S.J., R.H.F., J.L.C.) - all in California; Emory University School of Medicine and Children's Healthcare of Atlanta, Atlanta (E.I.F.); Endocrinologie Pédiatrique, Centre de Référence Maladies Endocriniennes Rares de la Croissance et du Développement, Hôpital Universitaire Robert-Debré, Groupe Hospitalo-Universitaire de l'Assistance Publique-Hôpitaux de Paris Nord, and Université Paris Cité, Faculté de Santé, UFR de Médecine, Paris, and Université Paris-Saclay, INSERM Physiologie et Physiopathologie Endocriniennes, Le Kremlin-Bicêtre - all in France (L.M.); the University of Colorado School of Medicine, Children's Hospital Colorado, Aurora (N.J.N.); Pediatric Endocrinology, Hospital Universitario Vall d'Hebrón, Barcelona (M.C.); the University of Washington School of Medicine, Seattle Children's Hospital, Seattle (P.Y.F.); the Children's Hospital of Philadelphia, Philadelphia (M.G.V.); Cohen Children's Medical Center of NY, New Hyde Park, and the Donald and Barbara Zucker School of Medicine at Hofstra-Northwell, Hempstead - both in New York (P.W.S.); and the Departments of Pharmacology and Internal Medicine, Division of Metabolism, Endocrinology and Diabetes, University of Michigan Medical School, and the Endocrinology and Metabolism Section, Medicine Services, LTC Charles S. Kettles Veterans Affairs Medical Center - both in Ann Arbor (R.J.A.)
| | - Richard J Auchus
- From the University of Minnesota Medical School and College of Pharmacy, Minneapolis (K.S.); Children's Hospital Los Angeles and Keck School of Medicine of USC, Los Angeles (M.S.K.), the University of California at San Francisco, Benioff Children's Hospital, San Francisco (M.L.), and Neurocrine Biosciences, San Diego (G.B.G.R., E.R., G.S.J., R.H.F., J.L.C.) - all in California; Emory University School of Medicine and Children's Healthcare of Atlanta, Atlanta (E.I.F.); Endocrinologie Pédiatrique, Centre de Référence Maladies Endocriniennes Rares de la Croissance et du Développement, Hôpital Universitaire Robert-Debré, Groupe Hospitalo-Universitaire de l'Assistance Publique-Hôpitaux de Paris Nord, and Université Paris Cité, Faculté de Santé, UFR de Médecine, Paris, and Université Paris-Saclay, INSERM Physiologie et Physiopathologie Endocriniennes, Le Kremlin-Bicêtre - all in France (L.M.); the University of Colorado School of Medicine, Children's Hospital Colorado, Aurora (N.J.N.); Pediatric Endocrinology, Hospital Universitario Vall d'Hebrón, Barcelona (M.C.); the University of Washington School of Medicine, Seattle Children's Hospital, Seattle (P.Y.F.); the Children's Hospital of Philadelphia, Philadelphia (M.G.V.); Cohen Children's Medical Center of NY, New Hyde Park, and the Donald and Barbara Zucker School of Medicine at Hofstra-Northwell, Hempstead - both in New York (P.W.S.); and the Departments of Pharmacology and Internal Medicine, Division of Metabolism, Endocrinology and Diabetes, University of Michigan Medical School, and the Endocrinology and Metabolism Section, Medicine Services, LTC Charles S. Kettles Veterans Affairs Medical Center - both in Ann Arbor (R.J.A.)
| | - Gelliza B G Rosales
- From the University of Minnesota Medical School and College of Pharmacy, Minneapolis (K.S.); Children's Hospital Los Angeles and Keck School of Medicine of USC, Los Angeles (M.S.K.), the University of California at San Francisco, Benioff Children's Hospital, San Francisco (M.L.), and Neurocrine Biosciences, San Diego (G.B.G.R., E.R., G.S.J., R.H.F., J.L.C.) - all in California; Emory University School of Medicine and Children's Healthcare of Atlanta, Atlanta (E.I.F.); Endocrinologie Pédiatrique, Centre de Référence Maladies Endocriniennes Rares de la Croissance et du Développement, Hôpital Universitaire Robert-Debré, Groupe Hospitalo-Universitaire de l'Assistance Publique-Hôpitaux de Paris Nord, and Université Paris Cité, Faculté de Santé, UFR de Médecine, Paris, and Université Paris-Saclay, INSERM Physiologie et Physiopathologie Endocriniennes, Le Kremlin-Bicêtre - all in France (L.M.); the University of Colorado School of Medicine, Children's Hospital Colorado, Aurora (N.J.N.); Pediatric Endocrinology, Hospital Universitario Vall d'Hebrón, Barcelona (M.C.); the University of Washington School of Medicine, Seattle Children's Hospital, Seattle (P.Y.F.); the Children's Hospital of Philadelphia, Philadelphia (M.G.V.); Cohen Children's Medical Center of NY, New Hyde Park, and the Donald and Barbara Zucker School of Medicine at Hofstra-Northwell, Hempstead - both in New York (P.W.S.); and the Departments of Pharmacology and Internal Medicine, Division of Metabolism, Endocrinology and Diabetes, University of Michigan Medical School, and the Endocrinology and Metabolism Section, Medicine Services, LTC Charles S. Kettles Veterans Affairs Medical Center - both in Ann Arbor (R.J.A.)
| | - Eiry Roberts
- From the University of Minnesota Medical School and College of Pharmacy, Minneapolis (K.S.); Children's Hospital Los Angeles and Keck School of Medicine of USC, Los Angeles (M.S.K.), the University of California at San Francisco, Benioff Children's Hospital, San Francisco (M.L.), and Neurocrine Biosciences, San Diego (G.B.G.R., E.R., G.S.J., R.H.F., J.L.C.) - all in California; Emory University School of Medicine and Children's Healthcare of Atlanta, Atlanta (E.I.F.); Endocrinologie Pédiatrique, Centre de Référence Maladies Endocriniennes Rares de la Croissance et du Développement, Hôpital Universitaire Robert-Debré, Groupe Hospitalo-Universitaire de l'Assistance Publique-Hôpitaux de Paris Nord, and Université Paris Cité, Faculté de Santé, UFR de Médecine, Paris, and Université Paris-Saclay, INSERM Physiologie et Physiopathologie Endocriniennes, Le Kremlin-Bicêtre - all in France (L.M.); the University of Colorado School of Medicine, Children's Hospital Colorado, Aurora (N.J.N.); Pediatric Endocrinology, Hospital Universitario Vall d'Hebrón, Barcelona (M.C.); the University of Washington School of Medicine, Seattle Children's Hospital, Seattle (P.Y.F.); the Children's Hospital of Philadelphia, Philadelphia (M.G.V.); Cohen Children's Medical Center of NY, New Hyde Park, and the Donald and Barbara Zucker School of Medicine at Hofstra-Northwell, Hempstead - both in New York (P.W.S.); and the Departments of Pharmacology and Internal Medicine, Division of Metabolism, Endocrinology and Diabetes, University of Michigan Medical School, and the Endocrinology and Metabolism Section, Medicine Services, LTC Charles S. Kettles Veterans Affairs Medical Center - both in Ann Arbor (R.J.A.)
| | - George S Jeha
- From the University of Minnesota Medical School and College of Pharmacy, Minneapolis (K.S.); Children's Hospital Los Angeles and Keck School of Medicine of USC, Los Angeles (M.S.K.), the University of California at San Francisco, Benioff Children's Hospital, San Francisco (M.L.), and Neurocrine Biosciences, San Diego (G.B.G.R., E.R., G.S.J., R.H.F., J.L.C.) - all in California; Emory University School of Medicine and Children's Healthcare of Atlanta, Atlanta (E.I.F.); Endocrinologie Pédiatrique, Centre de Référence Maladies Endocriniennes Rares de la Croissance et du Développement, Hôpital Universitaire Robert-Debré, Groupe Hospitalo-Universitaire de l'Assistance Publique-Hôpitaux de Paris Nord, and Université Paris Cité, Faculté de Santé, UFR de Médecine, Paris, and Université Paris-Saclay, INSERM Physiologie et Physiopathologie Endocriniennes, Le Kremlin-Bicêtre - all in France (L.M.); the University of Colorado School of Medicine, Children's Hospital Colorado, Aurora (N.J.N.); Pediatric Endocrinology, Hospital Universitario Vall d'Hebrón, Barcelona (M.C.); the University of Washington School of Medicine, Seattle Children's Hospital, Seattle (P.Y.F.); the Children's Hospital of Philadelphia, Philadelphia (M.G.V.); Cohen Children's Medical Center of NY, New Hyde Park, and the Donald and Barbara Zucker School of Medicine at Hofstra-Northwell, Hempstead - both in New York (P.W.S.); and the Departments of Pharmacology and Internal Medicine, Division of Metabolism, Endocrinology and Diabetes, University of Michigan Medical School, and the Endocrinology and Metabolism Section, Medicine Services, LTC Charles S. Kettles Veterans Affairs Medical Center - both in Ann Arbor (R.J.A.)
| | - Robert H Farber
- From the University of Minnesota Medical School and College of Pharmacy, Minneapolis (K.S.); Children's Hospital Los Angeles and Keck School of Medicine of USC, Los Angeles (M.S.K.), the University of California at San Francisco, Benioff Children's Hospital, San Francisco (M.L.), and Neurocrine Biosciences, San Diego (G.B.G.R., E.R., G.S.J., R.H.F., J.L.C.) - all in California; Emory University School of Medicine and Children's Healthcare of Atlanta, Atlanta (E.I.F.); Endocrinologie Pédiatrique, Centre de Référence Maladies Endocriniennes Rares de la Croissance et du Développement, Hôpital Universitaire Robert-Debré, Groupe Hospitalo-Universitaire de l'Assistance Publique-Hôpitaux de Paris Nord, and Université Paris Cité, Faculté de Santé, UFR de Médecine, Paris, and Université Paris-Saclay, INSERM Physiologie et Physiopathologie Endocriniennes, Le Kremlin-Bicêtre - all in France (L.M.); the University of Colorado School of Medicine, Children's Hospital Colorado, Aurora (N.J.N.); Pediatric Endocrinology, Hospital Universitario Vall d'Hebrón, Barcelona (M.C.); the University of Washington School of Medicine, Seattle Children's Hospital, Seattle (P.Y.F.); the Children's Hospital of Philadelphia, Philadelphia (M.G.V.); Cohen Children's Medical Center of NY, New Hyde Park, and the Donald and Barbara Zucker School of Medicine at Hofstra-Northwell, Hempstead - both in New York (P.W.S.); and the Departments of Pharmacology and Internal Medicine, Division of Metabolism, Endocrinology and Diabetes, University of Michigan Medical School, and the Endocrinology and Metabolism Section, Medicine Services, LTC Charles S. Kettles Veterans Affairs Medical Center - both in Ann Arbor (R.J.A.)
| | - Jean L Chan
- From the University of Minnesota Medical School and College of Pharmacy, Minneapolis (K.S.); Children's Hospital Los Angeles and Keck School of Medicine of USC, Los Angeles (M.S.K.), the University of California at San Francisco, Benioff Children's Hospital, San Francisco (M.L.), and Neurocrine Biosciences, San Diego (G.B.G.R., E.R., G.S.J., R.H.F., J.L.C.) - all in California; Emory University School of Medicine and Children's Healthcare of Atlanta, Atlanta (E.I.F.); Endocrinologie Pédiatrique, Centre de Référence Maladies Endocriniennes Rares de la Croissance et du Développement, Hôpital Universitaire Robert-Debré, Groupe Hospitalo-Universitaire de l'Assistance Publique-Hôpitaux de Paris Nord, and Université Paris Cité, Faculté de Santé, UFR de Médecine, Paris, and Université Paris-Saclay, INSERM Physiologie et Physiopathologie Endocriniennes, Le Kremlin-Bicêtre - all in France (L.M.); the University of Colorado School of Medicine, Children's Hospital Colorado, Aurora (N.J.N.); Pediatric Endocrinology, Hospital Universitario Vall d'Hebrón, Barcelona (M.C.); the University of Washington School of Medicine, Seattle Children's Hospital, Seattle (P.Y.F.); the Children's Hospital of Philadelphia, Philadelphia (M.G.V.); Cohen Children's Medical Center of NY, New Hyde Park, and the Donald and Barbara Zucker School of Medicine at Hofstra-Northwell, Hempstead - both in New York (P.W.S.); and the Departments of Pharmacology and Internal Medicine, Division of Metabolism, Endocrinology and Diabetes, University of Michigan Medical School, and the Endocrinology and Metabolism Section, Medicine Services, LTC Charles S. Kettles Veterans Affairs Medical Center - both in Ann Arbor (R.J.A.)
| |
Collapse
|
4
|
Finkielstain GP, Rey RA. Challenges in managing disorders of sex development associated with adrenal dysfunction. Expert Rev Endocrinol Metab 2023; 18:427-439. [PMID: 37694439 DOI: 10.1080/17446651.2023.2256393] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/02/2023] [Accepted: 09/04/2023] [Indexed: 09/12/2023]
Abstract
INTRODUCTION Disorders of Sex Development (DSD) associated with adrenal dysfunction occur due to different defects in the proteins involved in gonadal and adrenal steroidogenesis. AREAS COVERED The deficiencies in 21-hydroxylase and 11β-hydroxylase lead to DSD in 46,XX patients, defects in StAR, P450scc, 17α-hydroxylase and 17,20-lyase lead to 46,XY DSD, and 3β-HSD2 and POR deficiencies cause both 46,XX and 46,XY DSD. Challenges in diagnosis arise from the low prevalence and the variability in serum steroid profiles. Replacement therapy with hydrocortisone and fludrocortisone helps to minimize life-threatening adrenal crises; however, availability is still an unresolved problem in many countries. Adverse health outcomes, due to the disease or its treatment, are common and include adult short stature, hypertension, osteoporosis, obesity, cardiometabolic risk, and reproductive health issues. Potential biomarkers to improve monitoring and novel treatment options that have been developed with the primary aim to decrease adrenal androgen production are promising tools to help improve the health and quality of life of these patients. EXPERT OPINION Steroid profiling by mass spectrometry and next-generation sequencing technologies represent useful tools for establishing an etiologic diagnosis and drive personalized management. Nonetheless, access to health care still remains an issue requiring urgent solutions in many resource-limited settings.
Collapse
Affiliation(s)
- Gabriela P Finkielstain
- Centro de Investigaciones Endocrinológicas "Dr. César Bergadá" (CEDIE), CONICET - FEI - División de Endocrinología, Hospital de Niños Ricardo Gutiérrez, Buenos Aires, Argentina
| | - Rodolfo A Rey
- Centro de Investigaciones Endocrinológicas "Dr. César Bergadá" (CEDIE), CONICET - FEI - División de Endocrinología, Hospital de Niños Ricardo Gutiérrez, Buenos Aires, Argentina
| |
Collapse
|
5
|
Sarafoglou K, Merke DP, Reisch N, Claahsen-van der Grinten H, Falhammar H, Auchus RJ. Interpretation of Steroid Biomarkers in 21-Hydroxylase Deficiency and Their Use in Disease Management. J Clin Endocrinol Metab 2023; 108:2154-2175. [PMID: 36950738 PMCID: PMC10438890 DOI: 10.1210/clinem/dgad134] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Accepted: 03/07/2023] [Indexed: 03/24/2023]
Abstract
The most common form of congenital adrenal hyperplasia is 21-hydroxylase deficiency (21OHD), which in the classic (severe) form occurs in roughly 1:16 000 newborns worldwide. Lifelong treatment consists of replacing cortisol and aldosterone deficiencies, and supraphysiological dosing schedules are typically employed to simultaneously attenuate production of adrenal-derived androgens. Glucocorticoid titration in 21OHD is challenging as it must balance the consequences of androgen excess vs those from chronic high glucocorticoid exposure, which are further complicated by interindividual variability in cortisol kinetics and glucocorticoid sensitivity. Clinical assessment and biochemical parameters are both used to guide therapy, but the specific purpose and goals of each biomarker vary with age and clinical context. Here we review the approach to medication titration for children and adults with classic 21OHD, with an emphasis on how to interpret adrenal biomarker values in guiding this process. In parallel, we illustrate how an understanding of the pathophysiologic and pharmacologic principles can be used to avoid and to correct complications of this disease and consequences of its management using existing treatment options.
Collapse
Affiliation(s)
- Kyriakie Sarafoglou
- Department of Pediatrics, Division of Pediatric Endocrinology, University of Minnesota Medical School, Minneapolis, MN 55454, USA
- Department of Experimental and Clinical Pharmacology, University of Minnesota College of Pharmacy, Minneapolis, MN 55455, USA
| | - Deborah P Merke
- Department of Pediatrics, National Institutes of Health Clinical Center, Bethesda, MD 20892, USA
- Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, MD, USA
| | - Nicole Reisch
- Medizinische Klinik and Poliklinik IV, Klinikum der Universität München, 80336 Munich, Germany
| | - Hedi Claahsen-van der Grinten
- Department of Pediatrics, Amalia Children's Hospital, Radboud University Medical Center, 6500 HB, Nijmegen, The Netherlands
| | - Henrik Falhammar
- Department of Molecular Medicine and Surgery, Karolinska Institutet, SE-17176, Stockholm, Sweden
- Department of Endocrinology, Karolinska University Hospital, SE-17176, Stockholm, Sweden
| | - Richard J Auchus
- Departments of Pharmacology and Internal Medicine, Division of Metabolism, Endocrinology and Diabetes, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| |
Collapse
|
6
|
Auer MK, Nordenström A, Lajic S, Reisch N. Congenital adrenal hyperplasia. Lancet 2023; 401:227-244. [PMID: 36502822 DOI: 10.1016/s0140-6736(22)01330-7] [Citation(s) in RCA: 22] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Revised: 05/17/2022] [Accepted: 06/13/2022] [Indexed: 12/13/2022]
Abstract
Congenital adrenal hyperplasia is a group of autosomal recessive disorders leading to multiple complex hormonal imbalances caused by various enzyme deficiencies in the adrenal steroidogenic pathway. The most common type of congenital adrenal hyperplasia is due to steroid 21-hydroxylase (21-OHase, henceforth 21OH) deficiency. The rare, classic (severe) form caused by 21OH deficiency is characterised by life-threatening adrenal crises and is the most common cause of atypical genitalia in neonates with 46,XX karyotype. After the introduction of life-saving hormone replacement therapy in the 1950s and neonatal screening programmes in many countries, nowadays neonatal survival rates in patients with congenital adrenal hyperplasia are high. However, disease-related mortality is increased and therapeutic management remains challenging, with multiple long-term complications related to treatment and disease affecting growth and development, metabolic and cardiovascular health, and fertility. Non-classic (mild) forms of congenital adrenal hyperplasia caused by 21OH deficiency are more common than the classic ones; they are detected clinically and primarily identified in female patients with hirsutism or impaired fertility. Novel treatment approaches are emerging with the aim of mimicking physiological circadian cortisol rhythm or to reduce adrenal hyperandrogenism independent of the suppressive effect of glucocorticoids.
Collapse
Affiliation(s)
- Matthias K Auer
- Medizinische Klinik IV, Klinikum der Universität München, Munich, Germany
| | - Anna Nordenström
- Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden; Division of Paediatrics, Unit for Paediatric Endocrinology and Metabolic Disorders, Karolinska University Hospital, Stockholm, Sweden
| | - Svetlana Lajic
- Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden; Division of Paediatrics, Unit for Paediatric Endocrinology and Metabolic Disorders, Karolinska University Hospital, Stockholm, Sweden
| | - Nicole Reisch
- Medizinische Klinik IV, Klinikum der Universität München, Munich, Germany.
| |
Collapse
|
7
|
Raftopoulou C, Abawi O, Sommer G, Binou M, Paltoglou G, Flück CE, van den Akker ELT, Charmandari E. Leukocyte Telomere Length in Children With Congenital Adrenal Hyperplasia. J Clin Endocrinol Metab 2023; 108:443-452. [PMID: 36181470 DOI: 10.1210/clinem/dgac560] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Revised: 09/21/2022] [Indexed: 01/20/2023]
Abstract
CONTEXT Exposure to chronic stress and hypercortisolism is associated with decreased leukocyte telomere length (LTL), a marker for biological aging and cardiovascular disease. Children with congenital adrenal hyperplasia (CAH) are treated with glucocorticoids. OBJECTIVE To investigate LTL in children with CAH. METHODS In this prospective observational cohort study, conducted at 4 academic pediatric endocrinology outpatient clinics, children with genetically confirmed CAH were assessed at 2 follow-up visits (mean 4.1 ± 0.7 months apart). At each visit, LTL was determined by quantitative real-time PCR. All subjects underwent detailed clinical and endocrinologic evaluation and were classified as undertreated, optimally treated, or overtreated, accordingly. The influence of clinical factors on LTL was investigated using linear mixed models adjusted for age, sex, and BMI-z. RESULTS We studied 76 patients, of whom 31 (41%) were girls, 63 (83%) had classic CAH, 67 (88%) received hydrocortisone, and 8 (11%) prednisolone. Median age at first visit was 12.0 years (IQR, 6.3-15.1), and median BMI-z was 0.51 (IQR, -0.12 to 1.43). LTL was shorter in patients with classic vs nonclassic CAH (-0.29, P = 0.012), in overtreated than in optimally treated patients (-0.07, P = 0.002), and patients receiving prednisolone compared with hydrocortisone (-0.34, P < 0.001). LTL was not associated with undertreatment or daily hydrocortisone-equivalent dose (P > 0.05). CONCLUSION LTL is shorter in patients with classic than nonclassic CAH, and in those who are overtreated with hydrocortisone or treated with long-acting glucocorticoids. These findings may be attributed to chronic exposure to supraphysiologic glucocorticoid concentrations and indicate that LTL may be used as a biomarker for monitoring glucocorticoid treatment.
Collapse
Affiliation(s)
- Christina Raftopoulou
- Division of Endocrinology, Metabolism and Diabetes, First Department of Pediatrics, National and Kapodistrian University of Athens Medical School, 'Aghia Sophia' Children's Hospital, Athens 11527, Greece
- Division of Endocrinology and Metabolism, Center for Clinical, Experimental Surgery and Translational Research, Biomedical Research Foundation of the Academy of Athens, Athens 11527, Greece
| | - Ozair Abawi
- Division of Endocrinology, Department of Pediatrics, Erasmus MC-Sophia Children's Hospital, University Medical Center Rotterdam, Rotterdam 3000 CA, The Netherlands
| | - Grit Sommer
- Pediatric Endocrinology, Diabetology and Metabolism, Department of Pediatrics, Bern and Department of BioMedical Research, University Hospital Inselspital, University of Bern, Bern 3010, Switzerland
| | - Maria Binou
- Division of Endocrinology, Metabolism and Diabetes, First Department of Pediatrics, National and Kapodistrian University of Athens Medical School, 'Aghia Sophia' Children's Hospital, Athens 11527, Greece
- Division of Endocrinology and Metabolism, Center for Clinical, Experimental Surgery and Translational Research, Biomedical Research Foundation of the Academy of Athens, Athens 11527, Greece
| | - George Paltoglou
- Division of Endocrinology, Metabolism and Diabetes, First Department of Pediatrics, National and Kapodistrian University of Athens Medical School, 'Aghia Sophia' Children's Hospital, Athens 11527, Greece
- Division of Endocrinology and Metabolism, Center for Clinical, Experimental Surgery and Translational Research, Biomedical Research Foundation of the Academy of Athens, Athens 11527, Greece
| | - Christa E Flück
- Pediatric Endocrinology, Diabetology and Metabolism, Department of Pediatrics, Bern and Department of BioMedical Research, University Hospital Inselspital, University of Bern, Bern 3010, Switzerland
| | - Erica L T van den Akker
- Division of Endocrinology, Department of Pediatrics, Erasmus MC-Sophia Children's Hospital, University Medical Center Rotterdam, Rotterdam 3000 CA, The Netherlands
| | - E Charmandari
- Division of Endocrinology, Metabolism and Diabetes, First Department of Pediatrics, National and Kapodistrian University of Athens Medical School, 'Aghia Sophia' Children's Hospital, Athens 11527, Greece
- Division of Endocrinology and Metabolism, Center for Clinical, Experimental Surgery and Translational Research, Biomedical Research Foundation of the Academy of Athens, Athens 11527, Greece
| |
Collapse
|
8
|
Lawrence N, Bacila I, Dawson J, Bryce J, Ali SR, van den Akker ELT, Bachega TASS, Baronio F, Birkebæk NH, Bonfig W, van der Grinten HC, Costa EC, de Vries L, Elsedfy H, Güven A, Hannema S, Iotova V, van der Kamp HJ, Clemente M, Lichiardopol CR, Milenkovic T, Neumann U, Nordenström A, Poyrazoğlu Ş, Probst‐Scheidegger U, De Sanctis L, Tadokoro‐Cuccaro R, Thankamony A, Vieites A, Yavaş Z, Faisal Ahmed S, Krone N. Analysis of therapy monitoring in the International Congenital Adrenal Hyperplasia Registry. Clin Endocrinol (Oxf) 2022; 97:551-561. [PMID: 35781728 PMCID: PMC9796837 DOI: 10.1111/cen.14796] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Revised: 06/28/2022] [Accepted: 06/29/2022] [Indexed: 01/07/2023]
Abstract
OBJECTIVE Congenital adrenal hyperplasia (CAH) requires exogenous steroid replacement. Treatment is commonly monitored by measuring 17-OH progesterone (17OHP) and androstenedione (D4). DESIGN Retrospective cohort study using real-world data to evaluate 17OHP and D4 in relation to hydrocortisone (HC) dose in CAH patients treated in 14 countries. PATIENTS Pseudonymized data from children with 21-hydroxylase deficiency (21OHD) recorded in the International CAH Registry. MEASUREMENTS Assessments between January 2000 and October 2020 in patients prescribed HC were reviewed to summarise biomarkers 17OHP and D4 and HC dose. Longitudinal assessment of measures was carried out using linear mixed-effects models (LMEM). RESULTS Cohort of 345 patients, 52.2% female, median age 4.3 years (interquartile range: 3.1-9.2) were taking a median 11.3 mg/m2 /day (8.6-14.4) of HC. Median 17OHP was 35.7 nmol/l (3.0-104.0). Median D4 under 12 years was 0 nmol/L (0-2.0) and above 12 years was 10.5 nmol/L (3.9-21.0). There were significant differences in biomarker values between centres (p < 0.05). Correlation between D4 and 17OHP was good in multiple regression with age (p < 0.001, R2 = 0.29). In longitudinal assessment, 17OHP levels did not change with age, whereas D4 levels increased with age (p < 0.001, R2 = 0.08). Neither biomarker varied directly with dose or weight (p > 0.05). Multivariate LMEM showed HC dose decreasing by 1.0 mg/m2 /day for every 1 point increase in weight standard deviation score. DISCUSSION Registry data show large variability in 17OHP and D4 between centres. 17OHP correlates with D4 well when accounting for age. Prescribed HC dose per body surface area decreased with weight gain.
Collapse
Affiliation(s)
- Neil Lawrence
- Department of Oncology and MetabolismUniversity of SheffieldSheffieldUK
- Sheffield Children's Hospital NHS Foundation TrustSheffieldUK
| | - Irina Bacila
- Department of Oncology and MetabolismUniversity of SheffieldSheffieldUK
| | - Jeremy Dawson
- Institute of Work Psychology, Management SchoolUniversity of SheffieldSheffieldUK
- School of Health and Related Research, University of SheffieldSheffieldUK
| | - Jillian Bryce
- Office for Rare ConditionsRoyal Hospital for Children & Queen Elizabeth University HospitalGlasgowUK
- Office for Rare ConditionsRoyal Hospital for Children & Queen Elizabeth University HospitalGlasgowUK
| | - Salma R. Ali
- Office for Rare ConditionsRoyal Hospital for Children & Queen Elizabeth University HospitalGlasgowUK
- Office for Rare ConditionsRoyal Hospital for Children & Queen Elizabeth University HospitalGlasgowUK
- Developmental Endocrinology Research GroupUniversity of GlasgowGlasgowUK
| | - Erica L. T. van den Akker
- Department of Pediatric Endocrinology, Sophia Children's HospitalErasmus Medical CentreRotterdamthe Netherlands
| | - Tânia A. S. S. Bachega
- Hormones and Molecular Genetics Laboratory LIM 42, Department of Internal MedicineUniversity of Sao PauloSao PauloBrazil
| | - Federico Baronio
- Department of Medical and Surgical Sciences, Pediatric Unit, Endo‐ERN Center for Rare Endocrine DiseasesS. Orsola‐Malpighi University HospitalBolognaItaly
| | | | - Walter Bonfig
- Department of PediatricsTechnical University MunichMunichGermany
- Department of PediatricsKlinikum Wels‐GrieskirchenWelsAustria
| | - Hedi C. van der Grinten
- Department of Pediatric EndocrinologyRadboud University Medical CentreNijmegenthe Netherlands
- Amalia Children's HospitalRadboud University Medical CentreNijmegenthe Netherlands
| | - Eduardo C. Costa
- Pediatric Surgery ServiceHospital de Clínicas de Porto AlegrePorto AlegreBrazil
| | - Liat de Vries
- Institute for Diabetes and EndocrinologySchneider's Children Medical Center of IsraelPetah‐TikvahIsrael
| | - Heba Elsedfy
- Pediatrics DepartmentAin Shams UniversityCairoEgypt
| | - Ayla Güven
- Baskent University Istanbul HospitalPediatric EndocrinologyIstanbulTurkey
| | - Sabine Hannema
- Department of Paediatric Endocrinology, Erasmus MC, Sophia Children's HospitalUniversity Medical Center RotterdamRotterdamthe Netherlands
- Department of PaediatricsLeiden University Medical CentreLeidenthe Netherlands
| | - Violeta Iotova
- Department of PaediatricsMedical University of VarnaVarnaBulgaria
| | - Hetty J. van der Kamp
- Pediatric Endocrinology Wilhelmina Children's HospitalUniversity Medical Centre UtrechtUtrechtthe Netherlands
| | - María Clemente
- Paediatric Endocrinology, Hospital Universitario Vall d'HebronCIBER de Enfermedades Raras (CIBERER) ISCIIIBarcelonaSpain
| | | | - Tatjana Milenkovic
- Department of EndocrinologyInstitute for Mother and Child Healthcare of Serbia “Dr Vukan Čupić”BelgradeSerbia
| | - Uta Neumann
- Institute for Experimental Pediatric Endocrinology and Center for Chronically Sick Children, Charite‐UniversitätsmedizinBerlinGermany
| | - Ana Nordenström
- Department of Women's and Children's HealthKarolinska InstitutetStockholmSweden
- Department of Paediatric Endocrinology, Astrid Lindgren Children HospitalKarolinska University HospitalStockholmSweden
| | - Şukran Poyrazoğlu
- Istanbul Faculty of Medicine, Paediatric Endocrinology UnitIstanbul UniversityIstanbulTurkey
| | | | - Luisa De Sanctis
- Paediatric EndocrinologyRegina Margherita Children's HospitalTorinoItaly
- Department of Public Sciences and PediatricsUniversity of TorinoTorinoItaly
| | - Rieko Tadokoro‐Cuccaro
- Department of PediatricsUniversity of Cambridge, Cambridge, United Kingdom Biomedical CampusCambridgeUK
| | - Ajay Thankamony
- Department of PediatricsUniversity of Cambridge, Cambridge, United Kingdom Biomedical CampusCambridgeUK
| | - Ana Vieites
- Centro de Investigaciones Endocrinológicas (CEDIE‐CONICET), Hospital de Niños Ricardo GutiérrezBuenos AiresArgentina
| | - Zehra Yavaş
- Pediatric Endocrinology and DiabetesMarmara UniversityIstanbulTurkey
| | - Syed Faisal Ahmed
- Office for Rare ConditionsRoyal Hospital for Children & Queen Elizabeth University HospitalGlasgowUK
- Office for Rare ConditionsRoyal Hospital for Children & Queen Elizabeth University HospitalGlasgowUK
- Developmental Endocrinology Research GroupUniversity of GlasgowGlasgowUK
| | - Nils Krone
- Department of Oncology and MetabolismUniversity of SheffieldSheffieldUK
- Sheffield Children's Hospital NHS Foundation TrustSheffieldUK
- Department of Medicine IIIUniversity Hospital Carl Gustav Carus, Technische Universität DresdenDresdenGermany
| |
Collapse
|
9
|
Bacila I, Lawrence NR, Mahdi S, Alvi S, Cheetham TD, Crowne E, Das U, Dattani MT, Davies JH, Gevers E, Krone RE, Kyriakou A, Patel L, Randell T, Ryan FJ, Keevil B, Ahmed SF, Krone NP. Health status of children and young persons with congenital adrenal hyperplasia in the UK (CAH-UK): a cross-sectional multi-centre study. Eur J Endocrinol 2022; 187:543-553. [PMID: 36001026 PMCID: PMC9513639 DOI: 10.1530/eje-21-1109] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Accepted: 08/24/2022] [Indexed: 11/08/2022]
Abstract
OBJECTIVE There is limited knowledge on the onset of comorbidities in congenital adrenal hyperplasia (CAH) during childhood. We aimed to establish the health status of children with CAH in the UK. DESIGN AND METHODS This cross-sectional multicentre study involved 14 tertiary endocrine UK units, recruiting 101 patients aged 8-18 years with classic 21-hydroxylase deficiency and 83 controls. We analysed demographic, clinical and metabolic data, as well as psychological questionnaires (Strengths and Difficulties (SDQ), Paediatric Quality of Life (PedsQL)). RESULTS Patient height SDS in relation to mid-parental height decreased with age, indicating the discrepancy between height achieved and genetic potential height. Bone age was advanced in 40.5% patients, with a mean difference from the chronological age of 1.8 (±2.3) years. Patients were more frequently overweight (27%) or obese (22%) compared to controls (10.8% and 10.8%, respectively, P < 0.001). No consistent relationship between glucocorticoid dose and anthropometric measurements or hormonal biomarkers was detected. A small number of patients had raised total cholesterol (3.0%), low HDL (3.0%), raised LDL (7.0%) and triglycerides (5.0%). SDQ scores were within the 'high' and 'very high' categories of concern for 16.3% of patients. 'School functioning' was the lowest PedsQL scoring dimension with a median (interquartile range) of 70 (55-80), followed by 'emotional functioning' with a median of 75 (65-85). CONCLUSIONS Our results show an increased prevalence of problems with growth and weight gain in CAH children and suggest reduced quality of life. This highlights the urgent need to optimise management and monitoring strategies to improve long-term health outcomes.
Collapse
Affiliation(s)
- Irina Bacila
- Department of Oncology and Metabolism, University of Sheffield, Sheffield, UK
| | | | - Sundus Mahdi
- Department of Oncology and Metabolism, University of Sheffield, Sheffield, UK
| | | | | | - Elizabeth Crowne
- Bristol Royal Hospital for Children, University Hospitals Bristol Foundation Trust, Bristol, UK
| | - Urmi Das
- Alder Hey Children’s Hospital, Liverpool, UK
| | | | - Justin H Davies
- University Hospital Southampton, Southampton, UK
- Southampton, United Kingdom University of UK
| | - Evelien Gevers
- Centre for Endocrinology, William Harvey Research Institute, Queen Mary University London, London and Barts Health NHS Trust – The Royal London Hospital, London, UK
| | - Ruth E Krone
- Birmingham Women’s & Children’s Hospital, Birmingham, UK
| | - Andreas Kyriakou
- Developmental Endocrinology Research Group, University of Glasgow, Glasgow, UK
| | - Leena Patel
- Paediatric Endocrine Service, Royal Manchester Children’s Hospital, Manchester University NHS Foundation Trust, Manchester, UK
| | | | - Fiona J Ryan
- Oxford Children’s Hospital, Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| | - Brian Keevil
- Department of Biochemistry, Manchester University NHS Foundation Trust, Manchester, UK
| | - S Faisal Ahmed
- Developmental Endocrinology Research Group, University of Glasgow, Glasgow, UK
| | - Nils P Krone
- Department of Oncology and Metabolism, University of Sheffield, Sheffield, UK
- Correspondence should be addressed to N P Krone;
| |
Collapse
|
10
|
Ozdemir Dilek S, Turan I, Gurbuz F, Celiloglu C, Yuksel B. The utility of annual growth velocity standard deviation scores and measurements of biochemical parameters in long-term treatment monitoring of children with 21-hydroxylase deficiency. Hormones (Athens) 2022; 21:391-397. [PMID: 35243601 DOI: 10.1007/s42000-022-00354-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Accepted: 02/02/2022] [Indexed: 11/29/2022]
Abstract
PURPOSE This study aimed to investigate the utility of annual growth velocity (GV) standard deviation scores (SDSs) and compatibility and effectiveness of biochemical parameters in long-term treatment monitoring and management of 21-hydroxylase deficiency (21-OHD) in children. METHODS Fifty children with 21-OHD were included in this study, and the biochemical parameters obtained during 402 visits were retrospectively evaluated. The follow-up period was divided between two GV SDS groups (GV SDS < 2 and GV SDS ≥ 2) and compared with auxological, biochemical, and clinical findings. RESULTS Elevation of 17-hydroxyprogesterone (17-OHP) values was observed at 193/402 visits, and both adrenocorticotropic hormone (ACTH) and total testosterone (tT) were observed at 53 of 193 (27.5%) visits. The calculated cut-off value for 17-OHP was > 4.3 ng/ml, with a sensitivity of 85.48% and specificity of 37.59% in the GV SDS ≥ 2 group. In the GV SDS ≥ 2 group, the corrected final height SDS (cFH SDS) was lower, and the delta height was higher than in the GV SDS < 2 group (p = 0.005 and p = 0.008, respectively). Linear regression analysis of the GV SDSs revealed that 17-OHP values and the hydrocortisone dose (mg/m2) were affected (β = 0.037, p = 0.035, and β = - 0.147, p = 0.001, respectively). CONCLUSIONS Annual GV was critical in the final height (FH) of children with 21-OHD. However, we observed inconsistency between the biochemical parameters in the follow-ups, and there were difficulties in evaluating these markers. Therefore, annual GV SDSs and biochemical findings should be used together in patients with 21-OHD at follow-ups.
Collapse
Affiliation(s)
- Semine Ozdemir Dilek
- Department of Pediatrics, Division of Pediatric Endocrinology, Cukurova University Faculty of Medicine, Adana, Turkey
| | - Ihsan Turan
- Department of Pediatrics, Division of Pediatric Endocrinology, Cukurova University Faculty of Medicine, Adana, Turkey.
| | - Fatih Gurbuz
- Department of Pediatrics, Division of Pediatric Endocrinology, Cukurova University Faculty of Medicine, Adana, Turkey
| | - Can Celiloglu
- Department of Pediatrics, Division of Pediatric Endocrinology, Cukurova University Faculty of Medicine, Adana, Turkey
| | - Bilgin Yuksel
- Department of Pediatrics, Division of Pediatric Endocrinology, Cukurova University Faculty of Medicine, Adana, Turkey.
| |
Collapse
|
11
|
Abstract
Treatment for congenital adrenal hyperplasia (CAH) was introduced in the 1950s following the discovery of the structure and function of adrenocortical hormones. Although major advances in molecular biology have delineated steroidogenic mechanisms and the genetics of CAH, management and treatment of this condition continue to present challenges. Management is complicated by a combination of comorbidities that arise from disease-related hormonal derangements and treatment-related adverse effects. The clinical outcomes of CAH can include life-threatening adrenal crises, altered growth and early puberty, and adverse effects on metabolic, cardiovascular, bone and reproductive health. Standard-of-care glucocorticoid formulations fall short of replicating the circadian rhythm of cortisol and controlling efficient adrenocorticotrophic hormone-driven adrenal androgen production. Adrenal-derived 11-oxygenated androgens have emerged as potential new biomarkers for CAH, as traditional biomarkers are subject to variability and are not adrenal-specific, contributing to management challenges. Multiple alternative treatment approaches are being developed with the aim of tailoring therapy for improved patient outcomes. This Review focuses on challenges and advances in the management and treatment of CAH due to 21-hydroxylase deficiency, the most common type of CAH. Furthermore, we examine new therapeutic developments, including treatments designed to replace cortisol in a physiological manner and adjunct agents intended to control excess androgens and thereby enable reductions in glucocorticoid doses.
Collapse
Affiliation(s)
- Ashwini Mallappa
- National Institutes of Health Clinical Center, Bethesda, MD, USA
| | - Deborah P Merke
- National Institutes of Health Clinical Center, Bethesda, MD, USA.
- Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, MD, USA.
| |
Collapse
|
12
|
Rushworth RL, Chrisp GL, Bownes S, Torpy DJ, Falhammar H. Adrenal crises in adolescents and young adults. Endocrine 2022; 77:1-10. [PMID: 35583847 PMCID: PMC9242908 DOI: 10.1007/s12020-022-03070-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Accepted: 05/03/2022] [Indexed: 12/19/2022]
Abstract
PURPOSE Review the literature concerning adrenal insufficiency (AI) and adrenal crisis (AC) in adolescents and young adults. METHODS Searches of PubMed identifying relevant reports up to March 2022. RESULTS AI is rare disorder that requires lifelong glucocorticoid replacement therapy and is associated with substantial morbidity and occasional mortality among adolescents and young adults. Aetiologies in this age group are more commonly congenital, with acquired causes, resulting from tumours in the hypothalamic-pituitary area and autoimmune adrenalitis among others, increasing with age. All patients with AI are at risk of AC, which have an estimated incidence of 6 to 8 ACs/100 patient years. Prevention of ACs includes use of educational interventions to achieve competency in dose escalation and parenteral glucocorticoid administration during times of physiological stress, such as an intercurrent infection. While the incidence of AI/AC in young children and adults has been documented, there are few studies focussed on the AC occurrence in adolescents and young adults with AI. This is despite the range of developmental, psychosocial, and structural changes that can interfere with chronic disease management during this important period of growth and development. CONCLUSION In this review, we examine the current state of knowledge of AC epidemiology in emerging adults; examine the causes of ACs in this age group; and suggest areas for further investigation that are aimed at reducing the incidence and health impact of ACs in these patients.
Collapse
Affiliation(s)
- R Louise Rushworth
- School of Medicine, Sydney, The University of Notre Dame, 160 Oxford St, Darlinghurst, NSW, 2010, Australia
| | - Georgina L Chrisp
- School of Medicine, Sydney, The University of Notre Dame, 160 Oxford St, Darlinghurst, NSW, 2010, Australia
| | - Suzannah Bownes
- School of Medicine, Sydney, The University of Notre Dame, 160 Oxford St, Darlinghurst, NSW, 2010, Australia
| | - David J Torpy
- Endocrine and Metabolic Unit, Royal Adelaide Hospital, Adelaide, SA, Australia
- University of Adelaide, Adelaide, SA, Australia
| | - Henrik Falhammar
- Department of Endocrinology, Karolinska University Hospital, SE-17176, Stockholm, Sweden.
- Department of Molecular Medicine and Surgery, Karolinska Institutet, SE-17176, Stockholm, Sweden.
| |
Collapse
|
13
|
Nordenström A, Falhammar H, Lajic S. Current and Novel Treatment Strategies in Children with Congenital Adrenal Hyperplasia. Horm Res Paediatr 2022; 96:560-572. [PMID: 35086098 DOI: 10.1159/000522260] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Accepted: 01/19/2022] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND The standard treatment for congenital adrenal hyperplasia (CAH) in children is still hydrocortisone. Improved strategies for timing of the dose during the day and the dose per square meter body surface area used in children of different ages and developmental phases have improved the situation and outcome for the patients. Neonatal screening enables an earlier diagnosis and initiation of treatment, prevents from adrenal crisis, and improves growth and development also for children with the less severe forms of CAH. SUMMARY This review describes the current treatment strategies for children with CAH and discusses some potential treatment options that have been developed with the primary aim to decrease the adrenal androgen production. Novel modified release glucocorticoid therapies are also discussed. KEY MESSAGES The long-term effects of the new adjunct therapies are unknown, and some are not suitable for use in children and adolescents. The effects of the new therapies on bone mineral density, gonadal functions, and long-term cognitive development are yet to be assessed. It is not known what levels of adrenal androgens are optimal for normal growth, puberty, and bone health. The basis of using glucocorticoids and mineralocorticoids in the treatment of CAH remains, and in some individuals, it may be beneficial to add therapies to reduce the androgen load during certain life stages.
Collapse
Affiliation(s)
- Anna Nordenström
- Pediatric Endocrinology Unit, Department of Women's and Children's Health, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Henrik Falhammar
- Department of Molecular Medicine and Surgery, Department of Endocrinology, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Svetlana Lajic
- Pediatric Endocrinology Unit, Department of Women's and Children's Health, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
14
|
Troger T, Sommer G, Lang-Muritano M, Konrad D, Kuhlmann B, Zumsteg U, Flück CE. Characteristics of Growth in Children With Classic Congenital Adrenal Hyperplasia due to 21-Hydroxylase Deficiency During Adrenarche and Beyond. J Clin Endocrinol Metab 2022; 107:e487-e499. [PMID: 34599587 PMCID: PMC8764343 DOI: 10.1210/clinem/dgab701] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2021] [Indexed: 11/19/2022]
Abstract
CONTEXT Patients with classic congenital adrenal hyperplasia (CAH) often do not achieve their full growth potential. Adrenarche may accelerate bone maturation and thereby result in decreased growth in CAH. OBJECTIVE The study aimed to analyze the impact of growth during adrenarche on final height of adequately treated classic CAH patients. METHODS This retrospective, multicenter study (4 academic pediatric endocrinology centers) included 41 patients with classical CAH, born 1990-2012. We assessed skeletal maturation (bone age), growth velocity, and (projected) adult height outcomes, and analyzed potential influencing factors, such as sex, genotype, and glucocorticoid therapy. RESULTS Patients with classic CAH were shorter than peers (-0.4 SDS ± 0.8 SD) and their parents (corrected final height -0.6 SDS ± 1.0 SD). Analysis of growth during adrenarche revealed 2 different growth patterns: patients with accelerating bone age (49%), and patients with nonaccelerating bone age relative to chronological age (BA-CA). Patients with accelerating BA-CA were taller than the normal population during adrenarche years (P = 0.001) and were predicted to achieve lower adult height SDS (-0.9 SDS [95% CI, -1.3; -0.5]) than nonaccelerating patients when assessed during adrenarche (0.2 SDS [95% CI, -0.3; 0.8]). Final adult height was similarly reduced in both accelerating and nonaccelerating BA-CA groups (-0.4 SDS [95% CI, -0.9; 0.1] vs -0.3 SDS [95% CI, [-0.8; 0.1]). CONCLUSION Patients with and without significant bone age advancement, and thus differing height prediction during adrenarche, showed similar (predicted) final height when reassessed during pubertal years. Bone age alone should not be used during adrenarche as clinical marker for metabolic control in CAH treatment.
Collapse
Affiliation(s)
- Tobias Troger
- Division of Pediatric Endocrinology, Diabetology and Metabolism, Department of Pediatrics, Inselspital, Bern University Hospital, University of Bern, 3010 Bern, Switzerland
- Department of Biomedical Research, University of Bern, 3010 Bern, Switzerland
| | - Grit Sommer
- Division of Pediatric Endocrinology, Diabetology and Metabolism, Department of Pediatrics, Inselspital, Bern University Hospital, University of Bern, 3010 Bern, Switzerland
- Department of Biomedical Research, University of Bern, 3010 Bern, Switzerland
| | - Mariarosaria Lang-Muritano
- Department of Pediatric Endocrinology and Diabetology and Children’s Research Center, University Children’s Hospital Zurich, University of Zurich, 8032 Zürich, Switzerland
| | - Daniel Konrad
- Department of Pediatric Endocrinology and Diabetology and Children’s Research Center, University Children’s Hospital Zurich, University of Zurich, 8032 Zürich, Switzerland
| | | | - Urs Zumsteg
- Pediatric Endocrinology and Diabetology, University Children’s Hospital Basel UKBB, University of Basel, 4056 Basel, Switzerland
| | - Christa E Flück
- Division of Pediatric Endocrinology, Diabetology and Metabolism, Department of Pediatrics, Inselspital, Bern University Hospital, University of Bern, 3010 Bern, Switzerland
- Department of Biomedical Research, University of Bern, 3010 Bern, Switzerland
- Correspondence: Christa E. Flück, MD, Pediatric Endocrinology, Diabetology and Metabolism, University Children’s Hospital, Freiburgstrasse 15/ C845, 3010 Bern, Switzerland.
| |
Collapse
|
15
|
Claahsen - van der Grinten HL, Speiser PW, Ahmed SF, Arlt W, Auchus RJ, Falhammar H, Flück CE, Guasti L, Huebner A, Kortmann BBM, Krone N, Merke DP, Miller WL, Nordenström A, Reisch N, Sandberg DE, Stikkelbroeck NMML, Touraine P, Utari A, Wudy SA, White PC. Congenital Adrenal Hyperplasia-Current Insights in Pathophysiology, Diagnostics, and Management. Endocr Rev 2022; 43:91-159. [PMID: 33961029 PMCID: PMC8755999 DOI: 10.1210/endrev/bnab016] [Citation(s) in RCA: 187] [Impact Index Per Article: 93.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2021] [Indexed: 11/19/2022]
Abstract
Congenital adrenal hyperplasia (CAH) is a group of autosomal recessive disorders affecting cortisol biosynthesis. Reduced activity of an enzyme required for cortisol production leads to chronic overstimulation of the adrenal cortex and accumulation of precursors proximal to the blocked enzymatic step. The most common form of CAH is caused by steroid 21-hydroxylase deficiency due to mutations in CYP21A2. Since the last publication summarizing CAH in Endocrine Reviews in 2000, there have been numerous new developments. These include more detailed understanding of steroidogenic pathways, refinements in neonatal screening, improved diagnostic measurements utilizing chromatography and mass spectrometry coupled with steroid profiling, and improved genotyping methods. Clinical trials of alternative medications and modes of delivery have been recently completed or are under way. Genetic and cell-based treatments are being explored. A large body of data concerning long-term outcomes in patients affected by CAH, including psychosexual well-being, has been enhanced by the establishment of disease registries. This review provides the reader with current insights in CAH with special attention to these new developments.
Collapse
Affiliation(s)
| | - Phyllis W Speiser
- Cohen Children’s Medical Center of NY, Feinstein Institute, Northwell Health, Zucker School of Medicine, New Hyde Park, NY 11040, USA
| | - S Faisal Ahmed
- Developmental Endocrinology Research Group, School of Medicine Dentistry & Nursing, University of Glasgow, Glasgow, UK
| | - Wiebke Arlt
- Institute of Metabolism and Systems Research (IMSR), College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
- Department of Endocrinology, Queen Elizabeth Hospital, University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
| | - Richard J Auchus
- Division of Metabolism, Endocrinology, and Diabetes, Departments of Internal Medicine and Pharmacology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Henrik Falhammar
- Department of Molecular Medicine and Surgery, Karolinska Intitutet, Stockholm, Sweden
- Department of Endocrinology, Karolinska University Hospital, Stockholm, Sweden
| | - Christa E Flück
- Pediatric Endocrinology, Diabetology and Metabolism, Inselspital, Bern University Hospital, University of Bern, 3010 Bern, Switzerland
| | - Leonardo Guasti
- Centre for Endocrinology, William Harvey Research Institute, Bart’s and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Angela Huebner
- Division of Paediatric Endocrinology and Diabetology, Department of Paediatrics, Universitätsklinikum Dresden, Technische Universität Dresden, Dresden, Germany
| | - Barbara B M Kortmann
- Radboud University Medical Centre, Amalia Childrens Hospital, Department of Pediatric Urology, Nijmegen, The Netherlands
| | - Nils Krone
- Department of Oncology and Metabolism, University of Sheffield, Sheffield, UK
- Department of Medicine III, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Deborah P Merke
- National Institutes of Health Clinical Center and the Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, MD 20892, USA
| | - Walter L Miller
- Department of Pediatrics, Center for Reproductive Sciences, and Institute for Human Genetics, University of California, San Francisco, CA 94143, USA
| | - Anna Nordenström
- Department of Women’s and Children’s Health, Karolinska Institutet, Stockholm, Sweden
- Pediatric Endocrinology, Karolinska University Hospital, Stockholm, Sweden
| | - Nicole Reisch
- Medizinische Klinik IV, Klinikum der Universität München, Munich, Germany
| | - David E Sandberg
- Department of Pediatrics, Susan B. Meister Child Health Evaluation and Research Center, University of Michigan, Ann Arbor, MI 48109, USA
| | | | - Philippe Touraine
- Department of Endocrinology and Reproductive Medicine, Center for Rare Endocrine Diseases of Growth and Development, Center for Rare Gynecological Diseases, Hôpital Pitié Salpêtrière, Sorbonne University Medicine, Paris, France
| | - Agustini Utari
- Division of Pediatric Endocrinology, Department of Pediatrics, Faculty of Medicine, Diponegoro University, Semarang, Indonesia
| | - Stefan A Wudy
- Steroid Research & Mass Spectrometry Unit, Laboratory of Translational Hormone Analytics, Division of Paediatric Endocrinology & Diabetology, Justus Liebig University, Giessen, Germany
| | - Perrin C White
- Division of Pediatric Endocrinology, UT Southwestern Medical Center, Dallas TX 75390, USA
| |
Collapse
|
16
|
Adriaansen BPH, Schröder MAM, Span PN, Sweep FCGJ, van Herwaarden AE, Claahsen-van der Grinten HL. Challenges in treatment of patients with non-classic congenital adrenal hyperplasia. Front Endocrinol (Lausanne) 2022; 13:1064024. [PMID: 36578966 PMCID: PMC9791115 DOI: 10.3389/fendo.2022.1064024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Accepted: 11/25/2022] [Indexed: 12/14/2022] Open
Abstract
Congenital adrenal hyperplasia (CAH) due to 21α-hydroxylase deficiency (21OHD) or 11β-hydroxylase deficiency (11OHD) are congenital conditions with affected adrenal steroidogenesis. Patients with classic 21OHD and 11OHD have a (nearly) complete enzyme deficiency resulting in impaired cortisol synthesis. Elevated precursor steroids are shunted into the unaffected adrenal androgen synthesis pathway leading to elevated adrenal androgen concentrations in these patients. Classic patients are treated with glucocorticoid substitution to compensate for the low cortisol levels and to decrease elevated adrenal androgens levels via negative feedback on the pituitary gland. On the contrary, non-classic CAH (NCCAH) patients have more residual enzymatic activity and do generally not suffer from clinically relevant glucocorticoid deficiency. However, these patients may develop symptoms due to elevated adrenal androgen levels, which are most often less elevated compared to classic patients. Although glucocorticoid treatment can lower adrenal androgen production, the supraphysiological dosages also may have a negative impact on the cardiovascular system and bone health. Therefore, the benefit of glucocorticoid treatment is questionable. An individualized treatment plan is desirable as patients can present with various symptoms or may be asymptomatic. In this review, we discuss the advantages and disadvantages of different treatment options used in patients with NCCAH due to 21OHD and 11OHD.
Collapse
Affiliation(s)
- Bas P. H. Adriaansen
- Radboud Institute of Health Sciences, Department of Laboratory Medicine, Radboud University Medical Center, Nijmegen, Netherlands
- Department of Pediatric Endocrinology, Amalia Children’s Hospital, Radboud University Medical Center, Nijmegen, Netherlands
| | - Mariska A. M. Schröder
- Department of Pediatric Endocrinology, Amalia Children’s Hospital, Radboud University Medical Center, Nijmegen, Netherlands
| | - Paul N. Span
- Radiotherapy & OncoImmunology Laboratory, Radboud Institute of Molecular Life Sciences, Department of Radiation Oncology, Radboud University Medical Center, Nijmegen, Netherlands
| | - Fred C. G. J. Sweep
- Radboud Institute of Health Sciences, Department of Laboratory Medicine, Radboud University Medical Center, Nijmegen, Netherlands
| | - Antonius E. van Herwaarden
- Radboud Institute of Health Sciences, Department of Laboratory Medicine, Radboud University Medical Center, Nijmegen, Netherlands
| | - Hedi L. Claahsen-van der Grinten
- Department of Pediatric Endocrinology, Amalia Children’s Hospital, Radboud University Medical Center, Nijmegen, Netherlands
- *Correspondence: Hedi L. Claahsen-van der Grinten,
| |
Collapse
|
17
|
Hoyer-Kuhn H, Huebner A, Richter-Unruh A, Bettendorf M, Rohrer T, Kapelari K, Riedl S, Mohnike K, Dörr HG, Roehl FW, Fink K, Holl RW, Woelfle J. Hydrocortisone dosing in children with classic congenital adrenal hyperplasia: results of the German/Austrian registry. Endocr Connect 2021; 10:561-569. [PMID: 33909597 PMCID: PMC8183617 DOI: 10.1530/ec-21-0023] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Accepted: 04/28/2021] [Indexed: 12/26/2022]
Abstract
OBJECTIVE Treatment of classic congenital adrenal hyperplasia (CAH) is necessary to compensate for glucocorticoid/mineralocorticoid deficiencies and to suppress androgen excess. Hydrocortisone (HC) is preferred in growing children with classic CAH but recommendations regarding dosage/administration are inconsistent. The aim of this study was to evaluate HC dosing in children with CAH in relation to chronological age, sex, and phenotype based on a multicenter CAH registry. DESIGN The CAH registry was initiated in 1997 by the AQUAPE in Germany. On December 31st 2018, data from 1571 patients were included. METHODS A custom-made electronic health record software is used at the participating centers. Pseudonymized data are transferred for central analysis. Parameters were selected based on current guidelines. Descriptive analyses and linear regression models were implemented with SAS 9.4. RESULTS We identified 1288 patients on exclusive treatment with hydrocortisone three times daily (604 boys; median age 7.2 years; 817 salt-wasting phenotype, 471 simple-virilizing phenotype). The mean (lower-upper quartiles) daily HC dose (mg/m² body surface area) was 19.4 (18.9-19.8) for patients <3 months (n = 329), 15.0 (14.6-15.3) for age ≥3-12 months (n = 463), 14.0 (13.7-14.3) for age 1-5.9 years (n = 745), 14.2 (14.0-14.5) for age 6 years to puberty entry (n = 669), and 14.9 (14.6-15.2) during puberty to 18 years (n = 801). Fludrocortisone was administered in 74.1% of patients with a median daily dosage of 88.8 µg. CONCLUSION Our analyses showed that still a high proportion of children are treated with HC doses higher than recommended. This evaluation provides comprehensive information on nationwide hydrocortisone substitution dosages in children with CAH underlining the benefit of systematic data within a registry to assess daily practice.
Collapse
Affiliation(s)
- Heike Hoyer-Kuhn
- Department of Paediatrics, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
- Correspondence should be addressed to H Hoyer-Kuhn:
| | - Angela Huebner
- Department of Paediatrics, University Children’s Hospital Dresden, Dresden, Germany
| | | | | | - Tilman Rohrer
- University Children’s Hospital Homburg, Homburg, Germany
| | - Klaus Kapelari
- University Children’s Hospital Innsbruck, Innsbruck, Austria
| | - Stefan Riedl
- Department of Pediatric, Medical University of Vienna, Vienna, Austria
- St.Anna Kinderspital, Medical University of Vienna, Vienna, Austria
| | - Klaus Mohnike
- Department of Biometrics, Otto von Guericke Universität Magdeburg, Magdeburg, Sachsen-Anhalt, Germany
| | | | - Friedrich-Wilhelm Roehl
- Department of Biometrics, Otto von Guericke Universität Magdeburg, Magdeburg, Sachsen-Anhalt, Germany
| | - Katharina Fink
- Institute of Epidemiology and Medical Biometry, ZIBMT, University of Ulm, Ulm, Germany
| | - Reinhard W Holl
- Institute of Epidemiology and Medical Biometry, ZIBMT, University of Ulm, Ulm, Germany
| | | |
Collapse
|
18
|
Farkouh A, Baumgärtel C, Gottardi R, Hemetsberger M, Czejka M, Kautzky-Willer A. Sex-Related Differences in Drugs with Anti-Inflammatory Properties. J Clin Med 2021; 10:1441. [PMID: 33916167 PMCID: PMC8037587 DOI: 10.3390/jcm10071441] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Revised: 03/18/2021] [Accepted: 03/27/2021] [Indexed: 01/07/2023] Open
Abstract
There is increasing evidence of sex differences in the action of anti-inflammatory drugs, with women being at significantly higher risk of adverse effects. Nevertheless, clinicians' awareness of the implications of these sex differences on dosing and adverse event monitoring in routine practice is still in need of improvement. We reviewed the literature evaluating sex differences in terms of pharmacokinetics and pharmacodynamics of anti-inflammatory drugs. The anti-thrombotic activity of selective and non-selective COX-inhibitors tends to be stronger in men than women. Side effect profiles differ with regards to gastro-intestinal, renal and hepatic complications. Glucocorticosteroids were found to be more effective in men; women were more sensitive to corticosteroids when their oestradiol levels were high, a finding important for women taking hormonal contraception. TNF-alpha inhibitors have a longer half-life in men, leading to stronger immunosuppression and this a higher incidence of infections as side effects. Although research on sex differences in the effectiveness and safety of drugs is increasing, findings are often anecdotal and controversial. There is no systematic sex-differentiated reporting from clinical trials, and women are often under-represented. As personalized medicine is gaining in importance, sex, and gender aspects need to become integral parts of future research and policy making.
Collapse
Affiliation(s)
- André Farkouh
- Department of Pharmaceutical Sciences, University of Vienna, 1090 Vienna, Austria;
| | - Christoph Baumgärtel
- AGES Austrian Medicines and Medical Devices Agency and Austrian Federal Office for Safety in Health Care, 1200 Vienna, Austria;
| | - Roman Gottardi
- Vascular Surgery, MediClin Heart Institute Lahr/Baden, 77933 Lahr, Germany;
| | | | - Martin Czejka
- Department of Pharmaceutical Sciences, University of Vienna, 1090 Vienna, Austria;
| | - Alexandra Kautzky-Willer
- Gender Medicine Unit, Division of Endocrinology and Metabolism, Department of Internal Medicine III, Medical University of Vienna, 1090 Vienna, Austria;
| |
Collapse
|
19
|
Al-Kofahi M, Ahmed MA, Jaber MM, Tran TN, Willis BA, Zimmerman CL, Gonzalez-Bolanos MT, Brundage RC, Sarafoglou K. An integrated PK-PD model for cortisol and the 17-hydroxyprogesterone and androstenedione biomarkers in children with congenital adrenal hyperplasia. Br J Clin Pharmacol 2020; 87:1098-1110. [PMID: 32652643 PMCID: PMC9328191 DOI: 10.1111/bcp.14470] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Revised: 06/26/2020] [Accepted: 06/29/2020] [Indexed: 01/06/2023] Open
Abstract
Aims The aim of this study was to characterize the pharmacokinetic/pharmacodynamic relationships of cortisol and the adrenal biomarkers 17‐hydroxyprogesterone and androstenedione in children with congenital adrenal hyperplasia (CAH). Methods A nonlinear mixed‐effect modelling approach was used to analyse cortisol, 17‐hydroxyprogesterone and androstenedione concentrations obtained over 6 hours from children with CAH (n = 50). A circadian rhythm was evident and the model leveraged literature information on circadian rhythm in untreated children with CAH. Indirect response models were applied in which cortisol inhibited the production rate of all three compounds using an Imax model. Results Cortisol was characterized by a one‐compartment model with apparent clearance and volume of distribution estimated at 22.9 L/h/70 kg and 41.1 L/70 kg, respectively. The IC50 values of cortisol concentrations for cortisol, 17‐hydroxyprogesterone and androstenedione were estimated to be 1.36, 0.45 and 0.75 μg/dL, respectively. The inhibitory effect was found to be more potent on 17OHP than D4A, and the IC50 values were higher in salt‐wasting subjects than simple virilizers. Production rates of cortisol, 17‐hydroxyprogesterone and androstenedione were higher in simple‐virilizer subjects. Half‐lives of cortisol, 17‐hydroxyprogesterone and androstenedione were 60, 47 and 77 minutes, respectively. Conclusion Rapidly changing biomarker responses to cortisol concentrations highlight that single measurements provide volatile information about a child's disease control. Our model closely captured observed cortisol, 17‐hydroxyprogesterone and androstenedione concentrations. It can be used to predict concentrations over 24 hours and allows many novel exposure metrics to be calculated, e.g., AUC, AUC‐above‐threshold, time‐within‐range, etc. Our long‐range goal is to uncover dose–exposure–outcome relationships that clinicians can use in adjusting hydrocortisone dose and timing.
Collapse
Affiliation(s)
- Mahmoud Al-Kofahi
- Department of Experimental and Clinical Pharmacology, University of Minnesota, Minneapolis, MN, USA
| | - Mariam A Ahmed
- Department of Experimental and Clinical Pharmacology, University of Minnesota, Minneapolis, MN, USA.,College of Pharmacy, Helwan University, Egypt
| | - Mutaz M Jaber
- Department of Experimental and Clinical Pharmacology, University of Minnesota, Minneapolis, MN, USA
| | - Thang N Tran
- Department of Experimental and Clinical Pharmacology, University of Minnesota, Minneapolis, MN, USA
| | - Brian A Willis
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN, USA
| | - Cheryl L Zimmerman
- Department of Experimental and Clinical Pharmacology, University of Minnesota, Minneapolis, MN, USA
| | - Maria T Gonzalez-Bolanos
- Department of Pediatrics, University of Minnesota Masonic Children's Hospital, Minneapolis, MN, USA
| | - Richard C Brundage
- Department of Experimental and Clinical Pharmacology, University of Minnesota, Minneapolis, MN, USA
| | - Kyriakie Sarafoglou
- Department of Experimental and Clinical Pharmacology, University of Minnesota, Minneapolis, MN, USA.,Department of Pediatrics, University of Minnesota Masonic Children's Hospital, Minneapolis, MN, USA
| |
Collapse
|
20
|
Arafah BM. Perioperative Glucocorticoid Therapy for Patients with Adrenal Insufficiency: Dosing Based on Pharmacokinetic Data. J Clin Endocrinol Metab 2020; 105:5717686. [PMID: 31996925 DOI: 10.1210/clinem/dgaa042] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Accepted: 01/28/2020] [Indexed: 01/25/2023]
Abstract
BACKGROUND Perioperative glucocorticoid therapy for patients with adrenal insufficiency (AI) is currently based on anecdotal reports, without supporting pharmacokinetic data. METHODS We determined the half-life, clearance, and volume of distribution of 2 consecutive intravenously (IV)-administered doses of hydrocortisone (15 or 25 mg every 6 hours) to 22 dexamethasone-suppressed healthy individuals and used the data to develop a novel protocol to treat 68 patients with AI who required surgical procedures. Patients received 20 mg of hydrocortisone orally 2 to 4 hours before intubation and were started on 25 mg of IV hydrocortisone every 6 hours for 24 hours and 15 mg every 6 hours during the second day. Nadir cortisol concentrations were repeatedly measured during that period. RESULTS In healthy individuals, cortisol half-life was longer when the higher hydrocortisone dose was administered (2.02 ± 0.15 vs 1.81 ± 0.11 hours; P < 0.01), and in patients with AI, the half-life was longer than in healthy individuals given the same hydrocortisone dose. In both populations, the cortisol half-life increased further with the second hormone injection. Prolongation of cortisol half-life was due to decreased hydrocortisone clearance and an increase in its volume of distribution. Nadir cortisol levels determined throughout the 48 postoperative hours were within the range of values and often exceeded those observed perioperatively in patients without adrenal dysfunction. CONCLUSIONS Cortisol pharmacokinetics are altered in the postoperative period and indicate that lower doses of hydrocortisone can be safely administered to patients with AI undergoing major surgery. The findings of this investigation call into question the current practice of administering excessive glucocorticoid supplementation during stress.
Collapse
Affiliation(s)
- Baha M Arafah
- Division of Clinical and Molecular Endocrinology, Cleveland Medical Center and Case Western Reserve University, Cleveland, Ohio
| |
Collapse
|
21
|
Travers S, Bouvattier C, Fagart J, Martinerie L, Viengchareun S, Pussard E, Lombès M. Interaction between accumulated 21-deoxysteroids and mineralocorticoid signaling in 21-hydroxylase deficiency. Am J Physiol Endocrinol Metab 2020; 318:E102-E110. [PMID: 31821037 DOI: 10.1152/ajpendo.00368.2019] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
21-Hydroxylase deficiency (21OHD) is a rare genetic disorder in which salt-wasting syndrome occurs in 75% of cases, due to inability to synthesize cortisol and aldosterone. Recent mass spectrometry progress allowed identification of 21-deoxysteroids, i.e., 17-hydroxyprogesterone (17OHP), 21-deoxycortisol (21DF), and 21-deoxycorticosterone (21DB). We hypothesized that they may interfere with mineralocorticoid signaling and fludrocortisone therapy in patients with congenital adrenal hyperplasia (CAH) without effective glucocorticoid replacement and ACTH suppression. Our goal was to quantify circulating 21-deoxysteroids in a pediatric cohort with CAH related to 21OHD and to examine their impact on mineralocorticoid receptor (MR) activation. Twenty-nine patients with salt-wasting phenotype were classified in two groups according to their therapeutic control. During routine follow-up, 17OHP, 21DF, 21DB, and cortisol levels were quantified by liquid chromatography with tandem mass spectrometry before hydrocortisone intake and 1 and 2.5 h following treatment administration. Luciferase reporter gene assays were performed on transfected HEK293T cells while in silico modeling examined structural interactions between these steroids within ligand-binding domain of MR. Plasma 17OHP, 21DF, and 21DB accumulate in uncontrolled patients reaching micromolar concentrations even after hydrocortisone intake. 21DF and 21DB act as partial MR agonists with antagonist features similar to 17OHP, consistent with altered anchoring to Asn770 and unfavorable contact with Ala773 in ligand-binding pocket of MR. Our results demonstrate a complex interaction between all accumulating 21-deoxysteroids in uncontrolled 21OHD patients and mineralocorticoid signaling and suggest that appropriate steroid profiling should optimize management and follow-up of such patients, as keeping those steroids to low plasma levels should attest therapeutic efficacy and prevent interference with MR signaling.
Collapse
Affiliation(s)
- Simon Travers
- Institut National de la Santé et de la Recherche Médicale, Le Kremlin-Bicêtre, France
- Fac Med Paris-Sud, Université Paris-Sud, Université Paris Saclay, Le Kremlin-Bicêtre, France
- Département d'Endocrinologie Pédiatrique, Hôpital de Bicêtre, Hôpitaux Universitaires Paris Sud, Assistance Publique-Hôpitaux de Paris, Le Kremlin Bicêtre, France
| | - Claire Bouvattier
- Centre de Référence des Maladies Rares du Développement Génital (DEVGEN), Le Kremlin Bicêtre, France
- Service d'Endocrinologie Pédiatrique, Hôpital Robert Debré, Assistance Publique Hôpitaux de Paris, Paris, France
| | - Jérôme Fagart
- Institut National de la Santé et de la Recherche Médicale, Le Kremlin-Bicêtre, France
- Fac Med Paris-Sud, Université Paris-Sud, Université Paris Saclay, Le Kremlin-Bicêtre, France
| | - Laetitia Martinerie
- Institut National de la Santé et de la Recherche Médicale, Le Kremlin-Bicêtre, France
- Fac Med Paris-Sud, Université Paris-Sud, Université Paris Saclay, Le Kremlin-Bicêtre, France
- Université Paris Diderot, Sorbonne Paris Cité, Paris, France
| | - Say Viengchareun
- Institut National de la Santé et de la Recherche Médicale, Le Kremlin-Bicêtre, France
- Fac Med Paris-Sud, Université Paris-Sud, Université Paris Saclay, Le Kremlin-Bicêtre, France
| | - Eric Pussard
- Institut National de la Santé et de la Recherche Médicale, Le Kremlin-Bicêtre, France
- Fac Med Paris-Sud, Université Paris-Sud, Université Paris Saclay, Le Kremlin-Bicêtre, France
- Département d'Endocrinologie Pédiatrique, Hôpital de Bicêtre, Hôpitaux Universitaires Paris Sud, Assistance Publique-Hôpitaux de Paris, Le Kremlin Bicêtre, France
| | - Marc Lombès
- Institut National de la Santé et de la Recherche Médicale, Le Kremlin-Bicêtre, France
- Fac Med Paris-Sud, Université Paris-Sud, Université Paris Saclay, Le Kremlin-Bicêtre, France
| |
Collapse
|
22
|
Hindmarsh PC, Honour JW. Would Cortisol Measurements Be a Better Gauge of Hydrocortisone Replacement Therapy? Congenital Adrenal Hyperplasia as an Exemplar. Int J Endocrinol 2020; 2020:2470956. [PMID: 33299411 PMCID: PMC7701207 DOI: 10.1155/2020/2470956] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Revised: 07/27/2020] [Accepted: 09/24/2020] [Indexed: 11/18/2022] Open
Abstract
There is an increase in mortality and morbidity as well as poor quality of life in patients with congenital adrenal hyperplasia (CAH) and other causes of adrenal insufficiency. Glucocorticoid replacement therapy should aim to replace the missing cortisol as close as possible to the normal circadian rhythm using hydrocortisone. Dosing should be based on the individual's absorption and clearance of the drug. Adequacy of dosing should be checked using 24-hour profiles of plasma cortisol with samples drawn preferably every hour or at least every 2 hours. Measurement of cortisol should be the preferred method of assessing replacement therapy as it is over- and undertreatment with hydrocortisone, both of which can occur over a 24-hour period, which leads to the problems observed in patients with CAH and adrenal insufficiency.
Collapse
Affiliation(s)
- Peter C Hindmarsh
- Departments of Paediatrics, University College London Hospitals, London, UK
| | - John W Honour
- Institute for Women's Health, University College London, London, UK
| |
Collapse
|
23
|
Tajima T. Health problems of adolescent and adult patients with 21-hydroxylase deficiency. Clin Pediatr Endocrinol 2018; 27:203-213. [PMID: 30393437 PMCID: PMC6207803 DOI: 10.1297/cpe.27.203] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/16/2018] [Accepted: 07/02/2018] [Indexed: 12/30/2022] Open
Abstract
Twenty-one-hydroxylase deficiency (21-OHD) is one of the most common forms of congenital adrenal hyperplasias. Since the disease requires life-long steroid hormone replacement, transition from pediatric clinical care to adolescent and adult care is necessary. Recently, several studies have shown that morbidity and quality of life in adolescent and adult patients with 21-OHD are impaired by obesity, hypertension, diabetes mellitus, impaired glucose tolerance, dyslipidemia, and osteoporosis. In addition, excess adrenal androgen impairs fertility in both females and males. This mini review discusses the current health problems in adolescent and adult patients with 21-OHD and ways to prevent them.
Collapse
Affiliation(s)
- Toshihiro Tajima
- Jichi Medical University Children's Medical Center Tochigi, Tochigi, Japan
| |
Collapse
|
24
|
Speiser PW, Arlt W, Auchus RJ, Baskin LS, Conway GS, Merke DP, Meyer-Bahlburg HFL, Miller WL, Murad MH, Oberfield SE, White PC. Congenital Adrenal Hyperplasia Due to Steroid 21-Hydroxylase Deficiency: An Endocrine Society Clinical Practice Guideline. J Clin Endocrinol Metab 2018; 103:4043-4088. [PMID: 30272171 PMCID: PMC6456929 DOI: 10.1210/jc.2018-01865] [Citation(s) in RCA: 564] [Impact Index Per Article: 94.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/27/2018] [Accepted: 08/27/2018] [Indexed: 01/29/2023]
Abstract
Objective To update the congenital adrenal hyperplasia due to steroid 21-hydroxylase deficiency clinical practice guideline published by the Endocrine Society in 2010. Conclusions The writing committee presents updated best practice guidelines for the clinical management of congenital adrenal hyperplasia based on published evidence and expert opinion with added considerations for patient safety, quality of life, cost, and utilization.
Collapse
Affiliation(s)
- Phyllis W Speiser
- Cohen Children’s Medical Center of New York, New York, New York
- Zucker School of Medicine at Hofstra/Northwell, Hempstead, New York
| | - Wiebke Arlt
- University of Birmingham, Birmingham, United Kingdom
| | | | | | | | - Deborah P Merke
- National Institutes of Health Clinical Center, Bethesda, Maryland
- Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, Maryland
| | - Heino F L Meyer-Bahlburg
- New York State Psychiatric Institute, Vagelos College of Physicians & Surgeons of Columbia University, New York, New York
| | - Walter L Miller
- University of California San Francisco, San Francisco, California
| | - M Hassan Murad
- Mayo Clinic’s Evidence-Based Practice Center, Rochester, Minnesota
| | - Sharon E Oberfield
- NewYork–Presbyterian, Columbia University Medical Center, New York, New York
| | - Perrin C White
- University of Texas Southwestern Medical Center, Dallas, Texas
| |
Collapse
|
25
|
Peeters B, Meersseman P, Vander Perre S, Wouters PJ, Debaveye Y, Langouche L, Van den Berghe G. ACTH and cortisol responses to CRH in acute, subacute, and prolonged critical illness: a randomized, double-blind, placebo-controlled, crossover cohort study. Intensive Care Med 2018; 44:2048-2058. [PMID: 30374692 PMCID: PMC6280831 DOI: 10.1007/s00134-018-5427-y] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2018] [Accepted: 10/17/2018] [Indexed: 01/05/2023]
Abstract
Purpose Low plasma ACTH in critically ill patients may be explained by shock/inflammation-induced hypothalamus-pituitary damage or by feedback inhibition exerted by elevated plasma free cortisol. One can expect augmented/prolonged ACTH-responses to CRH injection with hypothalamic damage, immediately suppressed responses with pituitary damage, and delayed decreased responses in prolonged critical illness with feedback inhibition. Methods This randomized, double-blind, placebo-controlled crossover cohort study, compared ACTH responses to 100 µg IV CRH and placebo in 3 cohorts of 40 matched patients in the acute (ICU-day 3–6), subacute (ICU-day 7–16) or prolonged phase (ICU-day 17–28) of critical illness, with 20 demographically matched healthy subjects. CRH or placebo was injected in random order on two consecutive days. Blood was sampled repeatedly over 135 min and AUC responses to placebo were subtracted from those to CRH. Results Patients had normal mean ± SEM plasma ACTH concentrations (25.5 ± 1.6 versus 24.8 ± 3.6 pg/ml in healthy subjects, P = 0.54) but elevated free cortisol concentrations (3.11 ± 0.27 versus 0.58 ± 0.05 µg/dl in healthy subjects, P < 0.0001). The order of the CRH/placebo injections did not affect the ACTH responses, hence results were pooled. Patients in the acute phase of illness had normal mean ± SEM ACTH responses (5149 ± 848 pg/mL min versus 4120 ± 688 pg/mL min in healthy subjects; P = 0.77), whereas those in the subacute (2333 ± 387 pg/mL min, P = 0.01) and prolonged phases (2441 ± 685 pg/mL min, P = 0.001) were low, irrespective of sepsis/septic shock or risk of death. Conclusions Suppressed ACTH responses to CRH in the more prolonged phases, but not acute phase, of critical illness are compatible with feedback inhibition exerted by elevated free cortisol, rather than by cellular damage to hypothalamus and/or pituitary. Electronic supplementary material The online version of this article (10.1007/s00134-018-5427-y) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Bram Peeters
- Clinical Division and Laboratory of Intensive Care Medicine, Department of Cellular and Molecular Medicine, KU Leuven, Herestraat 49, 3000, Leuven, Belgium
| | - Philippe Meersseman
- Clinical Division and Laboratory of Intensive Care Medicine, Department of Cellular and Molecular Medicine, KU Leuven, Herestraat 49, 3000, Leuven, Belgium.,Medical Intensive Care Unit, Department of General Internal Medicine, UZ Leuven, Leuven, Belgium
| | - Sarah Vander Perre
- Clinical Division and Laboratory of Intensive Care Medicine, Department of Cellular and Molecular Medicine, KU Leuven, Herestraat 49, 3000, Leuven, Belgium
| | - Pieter J Wouters
- Clinical Division and Laboratory of Intensive Care Medicine, Department of Cellular and Molecular Medicine, KU Leuven, Herestraat 49, 3000, Leuven, Belgium
| | - Yves Debaveye
- Clinical Division and Laboratory of Intensive Care Medicine, Department of Cellular and Molecular Medicine, KU Leuven, Herestraat 49, 3000, Leuven, Belgium
| | - Lies Langouche
- Clinical Division and Laboratory of Intensive Care Medicine, Department of Cellular and Molecular Medicine, KU Leuven, Herestraat 49, 3000, Leuven, Belgium
| | - Greet Van den Berghe
- Clinical Division and Laboratory of Intensive Care Medicine, Department of Cellular and Molecular Medicine, KU Leuven, Herestraat 49, 3000, Leuven, Belgium.
| |
Collapse
|
26
|
Rushworth RL, Torpy DJ, Stratakis CA, Falhammar H. Adrenal Crises in Children: Perspectives and Research Directions. Horm Res Paediatr 2018; 89:341-351. [PMID: 29874655 DOI: 10.1159/000481660] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2017] [Accepted: 09/20/2017] [Indexed: 11/19/2022] Open
Abstract
Adrenal crises (AC) are life-threatening physiological disturbances that occur at a rate of 5-10/100 patient years in patients with adrenal insufficiency (AI). Despite their seriousness, there is a paucity of information on the epidemiology of AC events in the paediatric population specifically, as most investigations have focused on AI and ACs in adults. Improved surveillance of AC-related morbidity and mortality should improve the delineation of AC risk overall and among different subgroups of paediatric patients with AI. Valid incidence measures are essential for this purpose and also for the evaluation of interventions aimed at reducing adverse health outcomes from ACs. However, the absence of an agreed AC definition limits the potential benefit of research and surveillance in this area. While approaches to the treatment and prevention of ACs have much in common across the lifespan, there are important differences between children and adults with regards to the physiological, psychological, and social milieu in which these events occur. Education is considered to be an essential element of AC prevention but studies have shown that ACs occur even among well-educated patients, suggesting that new strategies may be needed. In this review, we examine the current knowledge regarding AC events in children with AI; assess the existing definitions of an AC and offer a new definition for use in research and the clinic; and suggest areas for further investigation that are aimed at reducing the incidence and health impact of ACs in the paediatric age group.
Collapse
Affiliation(s)
- R Louise Rushworth
- School of Medicine, Sydney, The University of Notre Dame, Darlinghurst, New South Wales, Australia
| | - David J Torpy
- Endocrine and Metabolic Unit, Royal Adelaide Hospital and University of Adelaide, Adelaide, South Australia, Australia
| | - Constantine A Stratakis
- Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland, USA
| | - Henrik Falhammar
- Department of Endocrinology, Metabolism and Diabetes, Karolinska University Hospital, Stockholm, Sweden.,Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden.,Menzies School of Health Research and Royal Darwin Hospital, Tiwi, Northwest Territories, Australia
| |
Collapse
|
27
|
Rushworth RL, Chrisp GL, Dean B, Falhammar H, Torpy DJ. Hospitalisation in Children with Adrenal Insufficiency and Hypopituitarism: Is There a Differential Burden between Boys and Girls and between Age Groups? Horm Res Paediatr 2018; 88:339-346. [PMID: 28898882 DOI: 10.1159/000479370] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/21/2017] [Accepted: 07/07/2017] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND/AIMS To determine the burden of hospitalisation in children with adrenal insufficiency (AI)/hypopituitarism in Australia. METHODS A retrospective study of Australian hospitalisation data. All admissions between 2001 and 2014 for patients aged 0-19 years with a principal diagnosis of AI/hypopituitarism were included. Denominator populations were extracted from national statistics datasets. RESULTS There were 3,779 admissions for treatment of AI/hypopituitarism in patients aged 0-19 years, corresponding to an average admission rate of 48.7 admissions/million/year. There were 470 (12.4%) admissions for an adrenal crisis (AC). Overall, admission for AI/hypopituitarism was comparable between the sexes. Admission rates for all AI, hypopituitarism, congenital adrenal hyperplasia (CAH), and "other and unspecified causes" of AI were highest among infants and decreased with age. Admissions for primary AI increased with age in both sexes. Males had significantly higher rates of admission for hypopituitarism. AC rates differed by both sex and age group. CONCLUSION This nationwide study of the epidemiology of hospital admissions for a principal diagnosis of AI/hypopituitarism shows that admissions generally decreased with age; males had higher rates of admission for hypopituitarism; females had higher rates of admission for CAH and "other and unspecified causes" of AI; and AC incidence varied by age and sex. Increased awareness of AI and AC prevention strategies may reduce some of these admissions.
Collapse
Affiliation(s)
- R Louise Rushworth
- School of Medicine, Sydney, The University of Notre Dame, Darlinghurst, New South Wales, Australia
| | - Georgina L Chrisp
- School of Medicine, Sydney, The University of Notre Dame, Darlinghurst, New South Wales, Australia
| | - Benjamin Dean
- School of Medicine, Sydney, The University of Notre Dame, Darlinghurst, New South Wales, Australia
| | - Henrik Falhammar
- Department of Endocrinology, Metabolism and Diabetes, Karolinska University Hospital, Stockholm, Sweden.,Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden.,Menzies School of Health Research and Royal Darwin Hospital, Tiwi, Northwest Territories, Australia
| | - David J Torpy
- Endocrine and Metabolic Unit, Royal Adelaide Hospital and University of Adelaide, Adelaide, South Australia, Australia
| |
Collapse
|
28
|
Madathilethu J, Roberts M, Peak M, Blair J, Prescott R, Ford JL. Content uniformity of quartered hydrocortisone tablets in comparison with mini-tablets for paediatric dosing. BMJ Paediatr Open 2018; 2:e000198. [PMID: 29637186 PMCID: PMC5843003 DOI: 10.1136/bmjpo-2017-000198] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2017] [Revised: 11/27/2017] [Accepted: 11/29/2017] [Indexed: 11/30/2022] Open
Abstract
OBJECTIVES Children requiring cortisol replacement therapy are often prescribed hydrocortisone doses of 2.5 mg, but as this is commercially unavailable 10 mg tablets, with functional break lines, are split commonly in an attempt to deliver the correct dose. This study aimed to determine the dose variation obtained from quartered hydrocortisone tablets when different operators performed the splitting procedure and to ascertain whether better uniformity could be attained from mini-tablets as an alternative formulation. METHODS Hydrocortisone 10 mg tablets were quartered by four different operators using a standard pill splitter. Hydrocortisone 2.5 mg mini-tablets (3 mm diameter) were formulated using a wet granulation method and manufactured using a high-speed rotary press simulator. The weight and content uniformity of the quartered tablets and mini-tablets were assessed according to pharmacopoeial standards. The physical strength and dissolution profiles of the mini-tablets were also determined. RESULTS More than half of all quartered 10 mg tablets were outside of the ±10% of the stated US Pharmacopoeia hydrocortisone content (mean 2.34 mg, SD 0.36, coefficient of variation (CV) 15.18%) and more than 40% of the quartered tablets were outside the European Pharmacopoeia weight variation. Robust mini-tablets (tensile strengths of >4 MPa) were produced successfully. The mini-tablets passed the pharmacopoeial weight and content uniformity requirements (mean 2.54 mg, SD 0.04, CV 1.72%) and drug release criteria during in vitro dissolution testing. CONCLUSION This study confirmed that quartering 10 mg hydrocortisone tablets produces unacceptable dose variations and that it is feasible to produce 3 mm mini-tablets containing more accurate doses for paediatric patients.
Collapse
Affiliation(s)
- Jude Madathilethu
- School of Pharmacy and Biomolecular Sciences, Liverpool John Moores University, Liverpool, UK
| | - Matthew Roberts
- School of Pharmacy and Biomolecular Sciences, Liverpool John Moores University, Liverpool, UK
| | - Matthew Peak
- Paediatric Medicines Research Unit, Alder Hey Children's NHS Foundation Trust, Liverpool, UK
| | - Joanne Blair
- Paediatric Medicines Research Unit, Alder Hey Children's NHS Foundation Trust, Liverpool, UK
| | - Rebecca Prescott
- School of Pharmacy and Biomolecular Sciences, Liverpool John Moores University, Liverpool, UK
| | - James L Ford
- School of Pharmacy and Biomolecular Sciences, Liverpool John Moores University, Liverpool, UK
| |
Collapse
|
29
|
Abstract
The congenital adrenal hyperplasias comprise a family of autosomal recessive disorders that disrupt adrenal steroidogenesis. The most common form is due to 21-hydroxylase deficiency associated with mutations in the 21-hydroxylase gene, which is located at chromosome 6p21. The clinical features associated with each disorder of adrenal steroidogenesis represent a clinical spectrum that reflect the consequences of the specific mutations. Treatment goals include normal linear growth velocity and "on-time" puberty in affected children. For adolescent and adult women, treatment goals include regularization of menses, prevention of progression of hirsutism, and preservation of fertility. For adolescent and adult men, prevention and early treatment of testicular adrenal rest tumors is beneficial. In this article key aspects regarding pathophysiology, diagnosis, and treatment of congenital adrenal hyperplasia are reviewed.
Collapse
Affiliation(s)
- Selma Feldman Witchel
- Division of Pediatric Endocrinology, Department of Pediatrics, Children's Hospital of Pittsburgh of UPMC, University of Pittsburgh, Pittsburgh, Pennsylvania.
| |
Collapse
|
30
|
Hodes A, Lodish MB, Tirosh A, Meyer J, Belyavskaya E, Lyssikatos C, Rosenberg K, Demidowich A, Swan J, Jonas N, Stratakis CA, Zilbermint M. Hair cortisol in the evaluation of Cushing syndrome. Endocrine 2017; 56:164-174. [PMID: 28194652 PMCID: PMC5437744 DOI: 10.1007/s12020-017-1231-7] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2016] [Accepted: 01/06/2017] [Indexed: 01/13/2023]
Abstract
PURPOSE Hair cortisol evaluation has been used to help detect patients with suspected Cushing syndrome. Our goal was to correlate segmental hair cortisol with biochemical testing in patients with Cushing syndrome and controls. This study was a prospective analysis of hair cortisol in confirmed Cushing syndrome cases over 16 months. METHODS Thirty-six subjects (26.5 ± 18.9 years, 75% female, and 75% Caucasian) were analyzed by diurnal serum cortisol, 24 h urinary free cortisol corrected for body surface area (UFC/BSA), and 24 h urinary 17-hydroxysteroids corrected for creatinine (17OHS/Cr). Thirty patients were diagnosed with Cushing syndrome, and six were defined as controls. 3-cm hair samples nearest to the scalp, cut into 1-cm segments (proximal, medial, and distal), were analyzed for cortisol by enzyme immunoassay and measured as pmol cortisol/g dry hair. Hair cortisol levels were compared with laboratory testing done within previous 2 months of the evaluation. RESULTS Proximal hair cortisol was higher in Cushing syndrome patients (266.6 ± 738.4 pmol/g) than control patients (38.9 ± 25.3 pmol/g) (p = 0.003). Proximal hair cortisol was highest of all segments in 25/36 (69%) patients. Among all subjects, proximal hair cortisol was strongly correlated with UFC/BSA (r = 0.5, p = 0.005), midnight serum cortisol (r = 0.4, p = 0.03), and 17OHS/Cr, which trended towards significance (r = 0.3, p = 0.06). CONCLUSIONS Among the three examined hair segments, proximal hair contained the highest cortisol levels and correlated the most with the initial biochemical tests for Cushing syndrome in our study. Further studies are needed to validate proximal hair cortisol in the diagnostic workup for Cushing syndrome.
Collapse
Affiliation(s)
- Aaron Hodes
- Section on Endocrinology and Genetics, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, 20892, USA
- Albert Einstein College of Medicine, Department of Radiology, Jacobi Medical Center, Bronx, NY, 10461, USA
| | - Maya B Lodish
- Section on Endocrinology and Genetics, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Amit Tirosh
- Section on Endocrinology and Genetics, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, 20892, USA
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, 6997801, Israel
| | - Jerrold Meyer
- Department of Psychological and Brain Sciences, University of Massachusetts, Amherst, MA, 01003, USA
| | - Elena Belyavskaya
- Section on Endocrinology and Genetics, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Charalampos Lyssikatos
- Section on Endocrinology and Genetics, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Kendra Rosenberg
- Department of Psychological and Brain Sciences, University of Massachusetts, Amherst, MA, 01003, USA
| | - Andrew Demidowich
- Section on Endocrinology and Genetics, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Jeremy Swan
- Section on Endocrinology and Genetics, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Nichole Jonas
- Section on Endocrinology and Genetics, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Constantine A Stratakis
- Section on Endocrinology and Genetics, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Mihail Zilbermint
- Section on Endocrinology and Genetics, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, 20892, USA.
- Johns Hopkins University School of Medicine, Division of Endocrinology, Diabetes, and Metabolism, Baltimore, MD, 21287, USA.
- Suburban Hospital, Bethesda, MD, 20814, USA.
| |
Collapse
|
31
|
Porter J, Blair J, Ross RJ. Is physiological glucocorticoid replacement important in children? Arch Dis Child 2017; 102:199-205. [PMID: 27582458 PMCID: PMC5284474 DOI: 10.1136/archdischild-2015-309538] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/24/2016] [Revised: 08/09/2016] [Accepted: 08/10/2016] [Indexed: 12/13/2022]
Abstract
Cortisol has a distinct circadian rhythm with low concentrations at night, rising in the early hours of the morning, peaking on waking and declining over the day to low concentrations in the evening. Loss of this circadian rhythm, as seen in jetlag and shift work, is associated with fatigue in the short term and diabetes and obesity in the medium to long term. Patients with adrenal insufficiency on current glucocorticoid replacement with hydrocortisone have unphysiological cortisol concentrations being low on waking and high after each dose of hydrocortisone. Patients with adrenal insufficiency complain of fatigue, a poor quality of life and there is evidence of poor health outcomes including obesity potentially related to glucocorticoid replacement. New technologies are being developed that deliver more physiological glucocorticoid replacement including hydrocortisone by subcutaneous pump, Plenadren, a once-daily modified-release hydrocortisone and Chronocort, a delayed and sustained absorption hydrocortisone formulation that replicates the overnight profile of cortisol. In this review, we summarise the evidence regarding physiological glucocorticoid replacement with a focus on relevance to paediatrics.
Collapse
Affiliation(s)
| | - Joanne Blair
- Department of Endocrinology, AlderHey Children's Hospital, Liverpool, UK
| | - Richard J Ross
- Diurnal Ltd, Cardiff, UK,Department of Endocrinology, The University of Sheffield, Sheffield, UK
| |
Collapse
|
32
|
Nella AA, Mallappa A, Perritt AF, Gounden V, Kumar P, Sinaii N, Daley LA, Ling A, Liu CY, Soldin SJ, Merke DP. A Phase 2 Study of Continuous Subcutaneous Hydrocortisone Infusion in Adults With Congenital Adrenal Hyperplasia. J Clin Endocrinol Metab 2016; 101:4690-4698. [PMID: 27680873 PMCID: PMC5155681 DOI: 10.1210/jc.2016-1916] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
CONTEXT Classic congenital adrenal hyperplasia (CAH) management remains challenging, given that supraphysiologic glucocorticoid doses are often needed to optimally suppress the ACTH-driven adrenal androgen overproduction. OBJECTIVE This study sought to approximate physiologic cortisol secretion via continuous subcutaneous hydrocortisone infusion (CSHI) and evaluate the safety and efficacy of CSHI in patients with difficult-to-treat CAH. DESIGN Eight adult patients with classic CAH participated in a single-center open-label phase I-II study comparing CSHI to conventional oral glucocorticoid treatment. All patients had elevated adrenal steroids and one or more comorbidities at study entry. Assessment while receiving conventional therapy at baseline and 6 months following CSHI included: 24-hour hormonal sampling, metabolic and radiologic evaluation, health-related quality-of-life (HRQoL), and fatigue questionnaires. MAIN OUTCOME MEASURES The ability of CSHI to approximate physiologic cortisol secretion and the percent of patients with 0700-hour 17-hydroxyprogesterone (17-OHP) ≤1200 ng/dL was measured. RESULTS CSHI approximated physiologic cortisol secretion. Compared with baseline, 6 months of CSHI resulted in decreased 0700-hour and 24-hour area under the curve 17-OHP, androstenedione, ACTH, and progesterone, increased osteocalcin, c-telopeptide and lean mass, and improved HRQoL (and SF-36 Vitality Score), and fatigue. One of three amenorrheic women resumed menses. One man had reduction of testicular adrenal rest tissue. CONCLUSIONS CSHI is a safe and well-tolerated modality of cortisol replacement that effectively approximates physiologic cortisol secretion in patients with classic CAH poorly controlled on conventional therapy. Improved adrenal steroid control and positive effects on HRQoL suggest that CSHI should be considered a treatment option for classic CAH. The long-term effect on established comorbidities requires further study.
Collapse
Affiliation(s)
- Aikaterini A Nella
- Eunice Kennedy Shriver National Institute of Child Health and Human Development (A.A.N. D.P.M.), National Institutes of Health, Bethesda, Maryland 20892; and National Institutes of Health Clinical Center (A.A.N., A.M., A.F.P., V.G., P.K., N.S., L.-A.D., A.L., C.-Y.L., S.J.S., D.P.M.), Bethesda, Maryland 20892
| | - Ashwini Mallappa
- Eunice Kennedy Shriver National Institute of Child Health and Human Development (A.A.N. D.P.M.), National Institutes of Health, Bethesda, Maryland 20892; and National Institutes of Health Clinical Center (A.A.N., A.M., A.F.P., V.G., P.K., N.S., L.-A.D., A.L., C.-Y.L., S.J.S., D.P.M.), Bethesda, Maryland 20892
| | - Ashley F Perritt
- Eunice Kennedy Shriver National Institute of Child Health and Human Development (A.A.N. D.P.M.), National Institutes of Health, Bethesda, Maryland 20892; and National Institutes of Health Clinical Center (A.A.N., A.M., A.F.P., V.G., P.K., N.S., L.-A.D., A.L., C.-Y.L., S.J.S., D.P.M.), Bethesda, Maryland 20892
| | - Verena Gounden
- Eunice Kennedy Shriver National Institute of Child Health and Human Development (A.A.N. D.P.M.), National Institutes of Health, Bethesda, Maryland 20892; and National Institutes of Health Clinical Center (A.A.N., A.M., A.F.P., V.G., P.K., N.S., L.-A.D., A.L., C.-Y.L., S.J.S., D.P.M.), Bethesda, Maryland 20892
| | - Parag Kumar
- Eunice Kennedy Shriver National Institute of Child Health and Human Development (A.A.N. D.P.M.), National Institutes of Health, Bethesda, Maryland 20892; and National Institutes of Health Clinical Center (A.A.N., A.M., A.F.P., V.G., P.K., N.S., L.-A.D., A.L., C.-Y.L., S.J.S., D.P.M.), Bethesda, Maryland 20892
| | - Ninet Sinaii
- Eunice Kennedy Shriver National Institute of Child Health and Human Development (A.A.N. D.P.M.), National Institutes of Health, Bethesda, Maryland 20892; and National Institutes of Health Clinical Center (A.A.N., A.M., A.F.P., V.G., P.K., N.S., L.-A.D., A.L., C.-Y.L., S.J.S., D.P.M.), Bethesda, Maryland 20892
| | - Lori-Ann Daley
- Eunice Kennedy Shriver National Institute of Child Health and Human Development (A.A.N. D.P.M.), National Institutes of Health, Bethesda, Maryland 20892; and National Institutes of Health Clinical Center (A.A.N., A.M., A.F.P., V.G., P.K., N.S., L.-A.D., A.L., C.-Y.L., S.J.S., D.P.M.), Bethesda, Maryland 20892
| | - Alexander Ling
- Eunice Kennedy Shriver National Institute of Child Health and Human Development (A.A.N. D.P.M.), National Institutes of Health, Bethesda, Maryland 20892; and National Institutes of Health Clinical Center (A.A.N., A.M., A.F.P., V.G., P.K., N.S., L.-A.D., A.L., C.-Y.L., S.J.S., D.P.M.), Bethesda, Maryland 20892
| | - Chia-Ying Liu
- Eunice Kennedy Shriver National Institute of Child Health and Human Development (A.A.N. D.P.M.), National Institutes of Health, Bethesda, Maryland 20892; and National Institutes of Health Clinical Center (A.A.N., A.M., A.F.P., V.G., P.K., N.S., L.-A.D., A.L., C.-Y.L., S.J.S., D.P.M.), Bethesda, Maryland 20892
| | - Steven J Soldin
- Eunice Kennedy Shriver National Institute of Child Health and Human Development (A.A.N. D.P.M.), National Institutes of Health, Bethesda, Maryland 20892; and National Institutes of Health Clinical Center (A.A.N., A.M., A.F.P., V.G., P.K., N.S., L.-A.D., A.L., C.-Y.L., S.J.S., D.P.M.), Bethesda, Maryland 20892
| | - Deborah P Merke
- Eunice Kennedy Shriver National Institute of Child Health and Human Development (A.A.N. D.P.M.), National Institutes of Health, Bethesda, Maryland 20892; and National Institutes of Health Clinical Center (A.A.N., A.M., A.F.P., V.G., P.K., N.S., L.-A.D., A.L., C.-Y.L., S.J.S., D.P.M.), Bethesda, Maryland 20892
| |
Collapse
|
33
|
Vajravelu ME, Tobolski J, Burrows E, Chilutti M, Xiao R, Bamba V, Willi S, Palladino A, Burnham JM, McCormack SE. Peak cortisol response to corticotropin-releasing hormone is associated with age and body size in children referred for clinical testing: a retrospective review. INTERNATIONAL JOURNAL OF PEDIATRIC ENDOCRINOLOGY 2015; 2015:22. [PMID: 26500680 PMCID: PMC4618529 DOI: 10.1186/s13633-015-0018-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/26/2015] [Accepted: 10/01/2015] [Indexed: 11/11/2022]
Abstract
Background Corticotropin-Releasing Hormone (CRH) testing is used to evaluate suspected adrenocorticotropic hormone (ACTH) deficiency, but the clinical characteristics that affect response in young children are incompletely understood. Our objective was to determine the effect of age and body size on cortisol response to CRH in children at risk for ACTH deficiency referred for clinical testing. Methods Retrospective, observational study of 297 children, ages 30 days – 18 years, undergoing initial, clinically indicated outpatient CRH stimulation testing at a tertiary referral center. All subjects received 1mcg/kg corticorelin per institutional protocol. Serial, timed ACTH and cortisol measurements were obtained. Patient demographic and clinical factors were abstracted from the medical record. Patients without full recorded anthropometric data, pubertal assessment, ACTH measurements, or clear indication for testing were excluded (number remaining = 222). Outcomes of interest were maximum cortisol after stimulation (peak) and cortisol rise from baseline (delta). Bivariable and multivariable linear regression analyses were used to assess the effects of age and size (weight, height, body mass index (BMI), body surface area (BSA), BMI z-score, and height z-score) on cortisol response while accounting for clinical covariates including sex, race/ethnicity, pubertal status, indication for testing, and time of testing. Results Subjects were 27 % female, with mean age of 8.9 years (SD 4.5); 75 % were pre-pubertal. Mean peak cortisol was 609.2 nmol/L (SD 213.0); mean delta cortisol was 404.2 nmol/L (SD 200.2). In separate multivariable models, weight, height, BSA and height z-score each remained independently negatively associated (p < 0.05) with peak and delta cortisol, controlling for indication of testing, baseline cortisol, and peak or delta ACTH, respectively. Age was negatively associated with peak but not delta cortisol in multivariable analysis. Conclusions Despite the use of a weight-based dosing protocol, both peak and delta cortisol response to CRH are negatively associated with several measures of body size in children referred for clinical testing, raising the question of whether alternate CRH dosing strategies or age- or size-based thresholds for adequate cortisol response should be considered in pediatric patients, or, alternatively, whether this finding reflects practice patterns followed when referring children for clinical testing. Electronic supplementary material The online version of this article (doi:10.1186/s13633-015-0018-y) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Mary Ellen Vajravelu
- Division of Endocrinology and Diabetes, The Children's Hospital of Philadelphia, 3401 Civic Center Blvd, Suite 11NW, Philadelphia, USA
| | - Jared Tobolski
- Division of Endocrinology and Diabetes, The Children's Hospital of Philadelphia, 3401 Civic Center Blvd, Suite 11NW, Philadelphia, USA
| | - Evanette Burrows
- Center for Biomedical Informatics, The Children's Hospital of Philadelphia, 3535 Market St, Philadelphia, PA 19104 USA
| | - Marianne Chilutti
- Center for Biomedical Informatics, The Children's Hospital of Philadelphia, 3535 Market St, Philadelphia, PA 19104 USA
| | - Rui Xiao
- Department of Biostatistics and Epidemiology, University of Pennsylvania, 635 Blockley Hall, 423 Guardian Drive, Philadelphia, PA 19104-6021 USA
| | - Vaneeta Bamba
- Division of Endocrinology and Diabetes, The Children's Hospital of Philadelphia, 3401 Civic Center Blvd, Suite 11NW, Philadelphia, USA
| | - Steven Willi
- Division of Endocrinology and Diabetes, The Children's Hospital of Philadelphia, 3401 Civic Center Blvd, Suite 11NW, Philadelphia, USA
| | - Andrew Palladino
- Global Innovative Pharma, Pfizer, Inc, 500 Arcola Road, Collegeville, PA 19426 USA
| | - Jon M Burnham
- The Children's Hospital of Philadelphia, Division of Rheumatology, 10103 Colket Building, 34th & Civic Center Blvd, Philadelphia, PA 19104 USA
| | - Shana E McCormack
- Division of Endocrinology and Diabetes, The Children's Hospital of Philadelphia, 3401 Civic Center Blvd, Suite 11NW, Philadelphia, USA
| |
Collapse
|
34
|
Ishii T, Anzo M, Adachi M, Onigata K, Kusuda S, Nagasaki K, Harada S, Horikawa R, Minagawa M, Minamitani K, Mizuno H, Yamakami Y, Fukushi M, Tajima T. Guidelines for diagnosis and treatment of 21-hydroxylase deficiency (2014 revision). Clin Pediatr Endocrinol 2015; 24:77-105. [PMID: 26594092 PMCID: PMC4639531 DOI: 10.1297/cpe.24.77] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2015] [Accepted: 03/10/2015] [Indexed: 11/07/2022] Open
Abstract
Purpose of developing the guidelines: The first guidelines for diagnosis and treatment of
21-hydroxylase deficiency (21-OHD) were published as a diagnostic handbook in Japan in
1989, with a focus on patients with severe disease. The “Guidelines for Treatment of
Congenital Adrenal Hyperplasia (21-Hydroxylase Deficiency) Found in Neonatal Mass
Screening (1999 revision)” published in 1999 were revised to include 21-OHD patients with
very mild or no clinical symptoms. Accumulation of cases and experience has subsequently
improved diagnosis and treatment of the disease. Based on these findings, the Mass
Screening Committee of the Japanese Society for Pediatric Endocrinology further revised
the guidelines for diagnosis and treatment. Target disease/conditions: 21-hydroxylase
deficiency. Users of the guidelines: Physician specialists in pediatric endocrinology,
pediatric specialists, referring pediatric practitioners, general physicians; and
patients.
Collapse
Affiliation(s)
| | | | | | - Tomohiro Ishii
- Department of Pediatrics, Keio University School of Medicine, Tokyo, Japan
| | - Makoto Anzo
- Department of Pediatrics, Kawasaki City Hospital, Kanagawa, Japan
| | - Masanori Adachi
- Department of Endocrinology and Metabolism, Kanagawa Children's Medical Center, Kanagawa, Japan
| | - Kazumichi Onigata
- Shimane University Hospital Postgraduate Clinical Training Center, Shimane, Japan
| | - Satoshi Kusuda
- Maternal and Perinatal Center, Tokyo Women's Medical University, Tokyo, Japan
| | - Keisuke Nagasaki
- Division of Pediatrics, Department of Homeostatic Regulation and Development, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Shohei Harada
- Division of Neonatal Screening, National Center for Child Health and Development, Tokyo, Japan
| | - Reiko Horikawa
- Department of Endocrinology and Metabolism, National Center for Child Health and Development, Tokyo, Japan
| | | | - Kanshi Minamitani
- Department of Pediatrics, Keio University School of Medicine, Tokyo, Japan
| | - Haruo Mizuno
- Departments of Pediatrics and Neonatology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Yuji Yamakami
- Kanagawa Health Service Association, Kanagawa, Japan
| | | | - Toshihiro Tajima
- Department of Pediatrics, Department of Pediatrics, Hokkaido University School of Medicine, Sapporo, Japan
| |
Collapse
|
35
|
Webb EA, Krone N. Current and novel approaches to children and young people with congenital adrenal hyperplasia and adrenal insufficiency. Best Pract Res Clin Endocrinol Metab 2015; 29:449-68. [PMID: 26051302 DOI: 10.1016/j.beem.2015.04.002] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Congenital adrenal hyperplasia (CAH) represents a group of autosomal recessive conditions leading to glucocorticoid deficiency. CAH is the most common cause of adrenal insufficiency (AI) in the paediatric population. The majority of the other forms of primary and secondary adrenal insufficiency are rare conditions. It is critical to establish the underlying aetiology of each specific condition as a wide range of additional health problems specific to the underlying disorder can be found. Following the introduction of life-saving glucocorticoid replacement sixty years ago, steroid hormone replacement regimes have been refined leading to significant reductions in glucocorticoid doses over the last two decades. These adjustments are made with the aim both of improving the current management of children and young persons and of reducing future health problems in adult life. However despite optimisation of existing glucocorticoid replacement regimens fail to mimic the physiologic circadian rhythm of glucocorticoid secretion, current efforts therefore focus on optimising replacement strategies. In addition, in recent years novel experimental therapies have been developed which target adrenal sex steroid synthesis in patients with CAH aiming to reduce co-morbidities associated with sex steroid excess. These developments will hopefully improve the health status and long-term outcomes in patients with congenital adrenal hyperplasia and adrenal insufficiency.
Collapse
Affiliation(s)
- Emma A Webb
- School of Clinical & Experimental Medicine, University of Birmingham, Institute of Biomedical Research, Birmingham B15 2TT, UK.
| | - Nils Krone
- School of Clinical & Experimental Medicine, University of Birmingham, Institute of Biomedical Research, Birmingham B15 2TT, UK.
| |
Collapse
|
36
|
Hindmarsh PC, Charmandari E. Variation in absorption and half-life of hydrocortisone influence plasma cortisol concentrations. Clin Endocrinol (Oxf) 2015; 82:557-61. [PMID: 25369980 DOI: 10.1111/cen.12653] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2014] [Revised: 09/26/2014] [Accepted: 10/30/2014] [Indexed: 11/26/2022]
Abstract
BACKGROUND Hydrocortisone therapy should be individualized in congenital adrenal hyperplasia (CAH) patients to avoid over and under replacement. We have assessed how differences in absorption and half-life of cortisol influence glucocorticoid exposure. PATIENTS AND METHODS Forty-eight patients (21 M) aged between 6·1 and 20·3 years with CAH due to CYP21A2 deficiency were studied. Each patient underwent a 24-h plasma cortisol profile with the morning dose used to calculate absorption parameters along with an intravenous (IV) hydrocortisone (15 mg/m(2) body surface area) bolus assessment of half-life. Parameters derived were maximum plasma concentration (Cmax ), time of maximum plasma concentration (tmax ), time to attaining plasma cortisol concentration <100 nmol/l and half-life of cortisol. RESULTS Mean half-life was 76·5 ± 5·2 (range 40-225·3) min, Cmax 780·7 ± 61·6 nmol/l and tmax 66·7 (range 20-118) min. Time taken to a plasma cortisol concentration less than 100 nmol/l was 289 (range 140-540) min. Those with a fast half-life and slow tmax took longest to reach a plasma cortisol concentration less than 100 nmol/l (380 ± 34·6 min), compared to those with a slow half-life and fast tmax (298 ± 34·8 min) and those with a fast half-life and fast tmax (249·5 ± 14·4 min) (One-way anovaF = 4·52; P = 0·009). CONCLUSIONS Both rate of absorption and half-life of cortisol in the circulation play important roles in determining overall exposure to oral glucocorticoid. Dose regimens need to incorporate estimates of these parameters into determining the optimum dosing schedule for individuals.
Collapse
Affiliation(s)
- Peter C Hindmarsh
- Developmental Endocrinology Research Group, UCL Institute of Child Health, London, UK
| | | |
Collapse
|
37
|
Interrelationships Among Cortisol, 17-Hydroxyprogesterone, and Androstenendione Exposures in the Management of Children With Congenital Adrenal Hyperplasia. J Investig Med 2015; 63:35-41. [DOI: 10.1097/jim.0000000000000121] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
38
|
Sarafoglou K, Gonzalez-Bolanos MT, Zimmerman CL, Boonstra T, Yaw Addo O, Brundage R. Comparison of cortisol exposures and pharmacodynamic adrenal steroid responses to hydrocortisone suspension vs. commercial tablets. J Clin Pharmacol 2014; 55:452-7. [PMID: 25385533 DOI: 10.1002/jcph.424] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2014] [Accepted: 11/04/2014] [Indexed: 11/10/2022]
Abstract
The Endocrine Society Clinical Practice Guidelines on congenital adrenal hyperplasia (CAH) recommend against using hydrocortisone suspension based on a study that examined a commercial suspension. Our objective was to examine the absorption of an extemporaneously prepared hydrocortisone suspension and compare it to tablets. Secondary objectives were to evaluate the 17-hydroxyprogesterone and androstenedione adrenal steroid responses. Using a parallel design, 34 children diagnosed with CAH received either suspension (n = 9; median age 1.8 years) or tablets (n = 25; median age 7.5 years). Patients were given their usual morning hydrocortisone formulation and dose; 12 serial blood samples were obtained and the area under the curve (AUC) was calculated. The mg/m(2) dose-normalized cortisol AUCs were no different in the suspension and tablet groups (P = ·06), nor was there a significant difference in the C(max) or T(max) (P = .08 and P = .41, respectively). Although there were no differences in the 17-hydroxyprogesterone change-from-baseline AUCs, baseline concentrations, or the nadir concentrations when comparing suspension and tablet formulations, the androstenedione values were significantly lower as expected in the younger aged suspension group. Our results offer compelling evidence that an extemporaneously prepared hydrocortisone suspension provides comparable cortisol exposures to commercially available tablet formulations in children and can be used to safely and effectively treat CAH.
Collapse
Affiliation(s)
- Kyriakie Sarafoglou
- Department of Pediatrics, University of Minnesota Children's Hospital, Minneapolis, MN, USA
| | | | | | | | | | | |
Collapse
|
39
|
Noppe G, van Rossum EFC, Vliegenthart J, Koper JW, van den Akker ELT. Elevated hair cortisol concentrations in children with adrenal insufficiency on hydrocortisone replacement therapy. Clin Endocrinol (Oxf) 2014; 81:820-5. [PMID: 25039686 DOI: 10.1111/cen.12551] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2014] [Revised: 04/20/2014] [Accepted: 07/07/2014] [Indexed: 11/28/2022]
Abstract
BACKGROUND Glucocorticoid replacement therapy in patients with adrenal insufficiency needs to be tailored to the individual patient based on body composition and clinical signs and symptoms as no objective method for assessment of treatment adequacy is available. Current treatment regimens are often not satisfactory, which is shown by the adverse metabolic profile and doubled mortality rates in treated adrenal insufficiency patients. Measurement of cortisol concentrations in hair reflect the long-term systemic cortisol exposure and may be of use in refinement of hydrocortisone treatment. OBJECTIVE We aimed to study whether long-term cortisol (hydrocortisone) levels, as measured in scalp hair, are similar in children with adrenal insufficiency and healthy children. MATERIAL AND METHODS We set up a case control study, measuring anthropometric characteristics and hair cortisol concentrations (HCC) in 54 hydrocortisone substituted children with adrenal insufficiency (AI patients) in the age of 4-18 years and 54 healthy children matched for gender and age. RESULTS Mean HCC were significantly higher in AI patients compared with healthy controls (mean 13·3 vs 8·2 pg/mg, P = 0·02). AI patients also had a higher BMI (P < 0·001) and waist circumference (WC) (P = 0·02). HCC was significantly associated with BMI (P = 0·002) and WC (P = 0·002). HCC explained 13% of the difference in BMI and 29% of the difference in WC between AI patients and controls. CONCLUSION Hydrocortisone-treated AI patients have increased HCC and adverse anthropometric characteristics compared with healthy controls. HCC measurement may be of value in identifying overtreatment and thereby improve hydrocortisone replacement therapy.
Collapse
Affiliation(s)
- G Noppe
- Division of Endocrinology, Department of Paediatrics, Sophia Children's Hospital, Erasmus MC, Rotterdam, The Netherlands; Division of Endocrinology, Department of Internal Medicine, Erasmus MC, Rotterdam, The Netherlands
| | | | | | | | | |
Collapse
|
40
|
Amr NH, Ahmed AY, Ibrahim YA. Carotid intima media thickness and other cardiovascular risk factors in children with congenital adrenal hyperplasia. J Endocrinol Invest 2014; 37:1001-8. [PMID: 25112902 DOI: 10.1007/s40618-014-0148-8] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2014] [Accepted: 07/28/2014] [Indexed: 12/21/2022]
Abstract
PURPOSE Patients with congenital adrenal hyperplasia (CAH) are at increased risk for cardiovascular disease due to many factors. The aim of this study is to investigate the presence of dyslipidemia, insulin resistance, and subclinical atherosclerosis as indicated by carotid intima media thickness in children with congenital adrenal hyperplasia. METHODS Thirty-two children with congenital adrenal hyperplasia (3-17 years) were compared with 32 healthy controls. All underwent anthropometric evaluation, measurement of fasting lipids, glucose, insulin, oral glucose tolerance test (OGTT), homeostasis model assessment for insulin resistance (HOMA-IR), and carotid intima media thickness (CIMT). RESULTS Fasting glucose, glucose at 30, 60, 90, and 120 min during OGTT were significantly higher in patients. HOMA-IR was also significantly higher in patients (p = 0.036). Patients had significantly higher CIMT (p = 0.003), and higher systolic blood pressure. (p = 0.04). No significant difference existed in lipid profile. Both systolic and diastolic blood pressures correlated with treatment duration (p = 0.002, p = 0.043, respectively). CONCLUSION Children with CAH are at increased risk of insulin resistance, glucose intolerance, early atherosclerosis, and cardiovascular disease. Screening of these patients at an early age is recommended.
Collapse
Affiliation(s)
- N H Amr
- Paediatrics Department, Ain Shams University, Cairo, Egypt,
| | | | | |
Collapse
|
41
|
Hindmarsh PC. The child with difficult to control Congenital Adrenal Hyperplasia: is there a place for continuous subcutaneous hydrocortisone therapy. Clin Endocrinol (Oxf) 2014; 81:15-8. [PMID: 24655023 DOI: 10.1111/cen.12453] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/02/2013] [Revised: 12/18/2013] [Accepted: 03/17/2014] [Indexed: 11/30/2022]
Abstract
The treatment of congenital adrenal hyperplasia requires administration of glucocorticoid and mineralocorticoid to replace the deficit resulting from the enzymatic block. During puberty, cortisol metabolism changes with an increase in clearance, which may lead to unstable control. This may require an increase in dosing frequency, and if this proves problematic, continuous subcutaneous hydrocortisone delivery using insulin pump technology can be valuable. The potential indications for such therapy along with some practical considerations are detailed in this Clinical Question report.
Collapse
Affiliation(s)
- Peter C Hindmarsh
- Developmental Endocrinology Research Group, UCL Institute of Child Health, London, UK
| |
Collapse
|
42
|
Abstract
Adrenal insufficiency is the clinical manifestation of deficient production or action of glucocorticoids, with or without deficiency also in mineralocorticoids and adrenal androgens. It is a life-threatening disorder that can result from primary adrenal failure or secondary adrenal disease due to impairment of the hypothalamic-pituitary axis. Prompt diagnosis and management are essential. The clinical manifestations of primary adrenal insufficiency result from deficiency of all adrenocortical hormones, but they can also include signs of other concurrent autoimmune conditions. In secondary or tertiary adrenal insufficiency, the clinical picture results from glucocorticoid deficiency only, but manifestations of the primary pathological disorder can also be present. The diagnostic investigation, although well established, can be challenging, especially in patients with secondary or tertiary adrenal insufficiency. We summarise knowledge at this time on the epidemiology, causal mechanisms, pathophysiology, clinical manifestations, diagnosis, and management of this disorder.
Collapse
Affiliation(s)
- Evangelia Charmandari
- Division of Endocrinology, Metabolism, and Diabetes, First Department of Pediatrics, University of Athens Medical School, Aghia Sophia Children's Hospital, Athens, Greece; Division of Endocrinology and Metabolism, Clinical Research Center, Biomedical Research Foundation of the Academy of Athens, Athens, Greece.
| | - Nicolas C Nicolaides
- Division of Endocrinology, Metabolism, and Diabetes, First Department of Pediatrics, University of Athens Medical School, Aghia Sophia Children's Hospital, Athens, Greece; Division of Endocrinology and Metabolism, Clinical Research Center, Biomedical Research Foundation of the Academy of Athens, Athens, Greece
| | - George P Chrousos
- Division of Endocrinology, Metabolism, and Diabetes, First Department of Pediatrics, University of Athens Medical School, Aghia Sophia Children's Hospital, Athens, Greece; Division of Endocrinology and Metabolism, Clinical Research Center, Biomedical Research Foundation of the Academy of Athens, Athens, Greece
| |
Collapse
|
43
|
Subbarayan A, Dattani MT, Peters CJ, Hindmarsh PC. Cardiovascular risk factors in children and adolescents with congenital adrenal hyperplasia due to 21-hydroxylase deficiency. Clin Endocrinol (Oxf) 2014; 80:471-7. [PMID: 23751160 PMCID: PMC4204515 DOI: 10.1111/cen.12265] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2013] [Revised: 03/26/2013] [Accepted: 06/04/2013] [Indexed: 11/28/2022]
Abstract
OBJECTIVE 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. METHODS A retrospective cross-sectional study of 107 children (39 m) with CAH aged 9·2 years (range 0·4-20·5 years). Anthropometric, systolic (SBP) and diastolic (DBP) blood pressure data were collected and expressed as standard deviation scores (SDS) using UK growth reference data and the Fourth Task Force data set, respectively. Fasting blood glucose with plasma insulin and lipids was measured, and insulin resistance (HOMA IR) calculated using the homoeostasis assessment model. RESULTS 23·6% (33% men; 18% women) of the cohort were obese (BMI SDS>2). BMI SDS was significantly higher (P < 0·001) when compared with the UK population. Nineteen (20·9%) of 91 patients (20% men; 21% women) had systolic hypertension and 8 [8·8% (8·6% men; 8·9% women)] had diastolic hypertension. Mean SBP [108 (SD 13·5)] mm Hg was significantly higher than the normal population (P < 0·001), but mean DBP was not (P = 0·07). Both SBP SDS and DBP SDS were not related to BMI SDS. 9·5% of the subjects had hyperlipidaemia, but HOMA IR was more favourable compared with the normal population. CONCLUSION Despite a reduction in steroid doses over the last decade, a number of children with CAH are still obese and hypertensive. Whether this reflects general population trends or indicates a need to further optimize treatment regimens remains to be determined.
Collapse
Affiliation(s)
- Anbezhil Subbarayan
- Department of Endocrinology, Great Ormond Street Hospital for ChildrenLondon, UK
| | - Mehul T Dattani
- Department of Endocrinology, Great Ormond Street Hospital for ChildrenLondon, UK
- Developmental Endocrinology Research Group, UCL Institute of Child HealthLondon, UK
| | - Catherine J Peters
- Department of Endocrinology, Great Ormond Street Hospital for ChildrenLondon, UK
| | - Peter C Hindmarsh
- Department of Endocrinology, Great Ormond Street Hospital for ChildrenLondon, UK
- Developmental Endocrinology Research Group, UCL Institute of Child HealthLondon, UK
- Correspondence: Peter Hindmarsh, Developmental Endocrinology Research Group, UCL Institute of Child Health, 30 Guilford Street, London WC1N 1EH, UK. Tel.: +44 207 405 9200 ext 5813; E-mail:
| |
Collapse
|
44
|
Falhammar H, Thorén M. Clinical outcomes in the management of congenital adrenal hyperplasia. Endocrine 2012; 41:355-73. [PMID: 22228497 DOI: 10.1007/s12020-011-9591-x] [Citation(s) in RCA: 72] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2011] [Accepted: 12/20/2011] [Indexed: 01/09/2023]
Abstract
Congenital adrenal hyperplasia (CAH) is a group of disorders affecting adrenal steroid synthesis. The most common form, 21-hydroxylase deficiency, leads to decreased production of cortisol and aldosterone with increased androgen secretion. In classic CAH glucocorticoid treatment can be life-saving, and provides symptom control, but must be given in an unphysiological manner with the risk of negative long-term outcomes. A late diagnosis or a severe phenotype or genotype has also a negative impact. These factors can result in impaired quality of life (QoL), increased cardiometabolic risk, short stature, osteoporosis and fractures, benign tumors, decreased fertility, and vocal problems. The prognosis has improved during the last decades, thanks to better clinical management and nowadays the most affected patients seem to have a good QoL. Very few patients above the age of 60 years have, however, been studied. Classifying patients according to genotype may give additional useful clinical information. The introduction of neonatal CAH screening may enhance long-term results. Monitoring of different risk factors and negative consequences should be done regularly in an attempt to improve clinical outcomes further.
Collapse
Affiliation(s)
- Henrik Falhammar
- Department of Endocrinology, Metabolism and Diabetes, Karolinska University Hospital, D2:04, 17176 Stockholm, Sweden,
| | | |
Collapse
|
45
|
Moreira RPP, Jorge AAL, Gomes LG, Kaupert LC, Massud Filho J, Mendonca BB, Bachega TASS. Pharmacogenetics of glucocorticoid replacement could optimize the treatment of congenital adrenal hyperplasia due to 21-hydroxylase deficiency. Clinics (Sao Paulo) 2011; 66:1361-6. [PMID: 21915484 PMCID: PMC3161212 DOI: 10.1590/s1807-59322011000800009] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/18/2011] [Accepted: 05/02/2011] [Indexed: 12/05/2022] Open
Abstract
INTRODUCTION 21-hydroxylase deficiency is an autosomal recessive disorder that causes glucocorticoid deficiency and increased androgen production. Treatment is based on glucocorticoid replacement; however, interindividual variability in the glucocorticoid dose required to achieve adequate hormonal control has been observed. OBJECTIVE The present study aimed to evaluate the association between polymorphic variants involved inglucocorticoid action and/or metabolism and the mean daily glucocorticoid dose in 21-hydroxylase deficiency patients. METHODS We evaluated 53 patients with classical forms of 21-hydroxylase deficiency who were receiving cortisone acetate. All patients were between four and six years of age and had normal androgen levels. RESULTS The P450 oxidoreductase A503V, HSD11B1 rs12086634, and CYP3A7*1C variants were found in 19%, 11.3% and 3.8% of the patients, respectively. The mean ± SD glucocorticoid dose in patients with the CYP3A7*1C and wild-type alleles was 13.9 ± 0.8 and 19.5 ± 3.2 mg/m²/d, respectively. We did not identify an association between the P450 oxidoreductase or HSD11B1 allelic variants and the mean glucocorticoid dose. CONCLUSION Patients carrying the CYP3A7*1C variant required a significantly lower mean glucocorticoid dose. Indeed, the CYP3A7*1C allele accounted for 20% of the variability in the cortisone acetate dose. The analysis of genes involved in glucocorticoid metabolism may be useful in the optimization of treatment of 21-hydroxylase deficiency.
Collapse
Affiliation(s)
- Ricardo P P Moreira
- Unidade de Endocrinologia do Desenvolvimento, Laboratório de Hormônios e Genética Molecular LIM 42, Disciplina de Endocrinologia da Faculdade de Medicina da Universidad, Universidade de São Paulo, São Paulo, SP, Brazil
| | | | | | | | | | | | | |
Collapse
|
46
|
Speiser PW. Medical treatment of classic and nonclassic congenital adrenal hyperplasia. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2011; 707:41-5. [PMID: 21691951 DOI: 10.1007/978-1-4419-8002-1_9] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Affiliation(s)
- Phyllis W Speiser
- Division of Pediatric Endocrinology, Cohen Children's Medical Center of New York, New Hyde Park, NY 11040, USA.
| |
Collapse
|
47
|
Speiser PW, Azziz R, Baskin LS, Ghizzoni L, Hensle TW, Merke DP, Meyer-Bahlburg HFL, Miller WL, Montori VM, Oberfield SE, Ritzen M, White PC. Congenital adrenal hyperplasia due to steroid 21-hydroxylase deficiency: an Endocrine Society clinical practice guideline. J Clin Endocrinol Metab 2010; 95:4133-60. [PMID: 20823466 PMCID: PMC2936060 DOI: 10.1210/jc.2009-2631] [Citation(s) in RCA: 659] [Impact Index Per Article: 47.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
OBJECTIVE We developed clinical practice guidelines for congenital adrenal hyperplasia (CAH). PARTICIPANTS The Task Force included a chair, selected by The Endocrine Society Clinical Guidelines Subcommittee (CGS), ten additional clinicians experienced in treating CAH, a methodologist, and a medical writer. Additional experts were also consulted. The authors received no corporate funding or remuneration. CONSENSUS PROCESS Consensus was guided by systematic reviews of evidence and discussions. The guidelines were reviewed and approved sequentially by The Endocrine Society's CGS and Clinical Affairs Core Committee, members responding to a web posting, and The Endocrine Society Council. At each stage, the Task Force incorporated changes in response to written comments. CONCLUSIONS We recommend universal newborn screening for severe steroid 21-hydroxylase deficiency followed by confirmatory tests. We recommend that prenatal treatment of CAH continue to be regarded as experimental. The diagnosis rests on clinical and hormonal data; genotyping is reserved for equivocal cases and genetic counseling. Glucocorticoid dosage should be minimized to avoid iatrogenic Cushing's syndrome. Mineralocorticoids and, in infants, supplemental sodium are recommended in classic CAH patients. We recommend against the routine use of experimental therapies to promote growth and delay puberty; we suggest patients avoid adrenalectomy. Surgical guidelines emphasize early single-stage genital repair for severely virilized girls, performed by experienced surgeons. Clinicians should consider patients' quality of life, consulting mental health professionals as appropriate. At the transition to adulthood, we recommend monitoring for potential complications of CAH. Finally, we recommend judicious use of medication during pregnancy and in symptomatic patients with nonclassic CAH.
Collapse
Affiliation(s)
- Phyllis W Speiser
- Cohen Children's Medical Center of New York and Hofstra University School of Medicine, New Hyde Park, New York 11040, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Verma S, Vanryzin C, Sinaii N, Kim MS, Nieman LK, Ravindran S, Calis KA, Arlt W, Ross RJ, Merke DP. A pharmacokinetic and pharmacodynamic study of delayed- and extended-release hydrocortisone (Chronocort) vs. conventional hydrocortisone (Cortef) in the treatment of congenital adrenal hyperplasia. Clin Endocrinol (Oxf) 2010; 72:441-7. [PMID: 19486026 PMCID: PMC2866132 DOI: 10.1111/j.1365-2265.2009.03636.x] [Citation(s) in RCA: 74] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
OBJECTIVE Existing glucocorticoid treatment for congenital adrenal hyperplasia (CAH) is suboptimal and nonphysiological. We compared hormonal profiles during therapy with a new modified-release hydrocortisone (MR-HC), Chronocort, to conventional hydrocortisone (HC), Cortef, in patients with CAH. DESIGN AND PATIENTS We conducted a Phase 2, open-label, crossover pharmacokinetic and pharmacodynamic study in 14 patients (out of whom seven were male subjects, age ranging from 17 to 55) with classic 21-hydroxylase deficiency. One week of thrice daily HC (10, 5 and 15 mg) was followed by 1 month of once daily MR-HC (30 mg at 22:00 hours). Twenty four-hour sampling of cortisol, 17-hydroxyprogesterone (17-OHP), androstenedione, and ACTH was performed at steady state. MEASUREMENTS The primary outcome measures were 8- and 24-h area under the curve (AUC) hormones and 08:00 hours 17-OHP. RESULTS Hydrocortisone therapy resulted in three cortisol peaks. A single cortisol peak occurred at approximately 06:00 hours on MR-HC. MR-HC resulted in significantly (P < 0.001) lower 24-h afternoon (12:00 to 20:00 hours), and night-time (20:00 to 04:00 hours) cortisol as compared with HC. From 04:00 to 12:00 hours, when physiological cortisol is highest, cortisol was higher on MR-HC than HC (P < 0.001). Patients on MR-HC had significantly (P < 0.05) higher afternoon (12:00 to 20:00 hours) 17-OHP, androstenedione and ACTH, but significantly (P = 0.025) lower 08:00 hours 17-OHP. No serious adverse events occurred. CONCLUSIONS Modified-release hydrocortisone represents a promising new treatment for CAH. Overnight adrenal androgens were well-controlled, but rose in the afternoon with once-daily dosing suggesting that a morning dose of glucocorticoid is needed. Further studies are needed to determine the optimal dosing regimen and long-term clinical outcome.
Collapse
Affiliation(s)
- Somya Verma
- Reproductive Biology and Medicine Branch, The Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, MD, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Bryan SM, Honour JW, 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; 94:3477-80. [PMID: 19567522 DOI: 10.1210/jc.2009-0630] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/12/2023]
Abstract
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.
Collapse
Affiliation(s)
- Sinead M Bryan
- Developmental Endocrinology Research Group, Institute of Child Health, University College London, 30 Guilford Street, London WC1N 1EH, United Kingdom
| | | | | |
Collapse
|
50
|
Riepe FG, Sippell WG. Recent advances in diagnosis, treatment, and outcome of congenital adrenal hyperplasia due to 21-hydroxylase deficiency. Rev Endocr Metab Disord 2007; 8:349-63. [PMID: 17885806 DOI: 10.1007/s11154-007-9053-1] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
Congenital adrenal hyperplasia (CAH) due to 21-hydroxylase deficiency (21-OHD) is an autosomal-recessive disease causing cortisol deficiency, aldosterone deficiency and hyperandrogenism. Diagnosis of 21-OHD is confirmed by steroid analysis in newborn screening or later on. Standard medical treatment consists of oral glucocorticoid and mineralocorticoid administration in order to suppress adrenal androgens and to compensate for adrenal steroid deficiencies. However, available treatment is far from ideal, and not much is known about the long-term outcome in CAH as trials in patients in adulthood or old age are rare. Here we briefly describe the pathophysiology, clinical picture, genetics and epidemiology of 21-OHD. This is followed by a comprehensive review of the recent advances in diagnosis, treatment and outcome. Novel insights have been gained in the fields of newborn screening, specific steroid measurement utilizing mass spectrometry, genetics, glucocorticoid stress dosing, additive medical therapy, prenatal treatment, side-effects of medical treatment, adrenomedullary involvement, metabolic morbidity, fertility and gender identity. However, many issues are still unresolved, and novel questions, which will have to be answered in the future, arise with every new finding.
Collapse
Affiliation(s)
- Felix G Riepe
- Division of Pediatric Endocrinology, Department of Pediatrics, Christian-Albrechts-Universität Kiel, Schwanenweg 20, 24105, Kiel, Germany.
| | | |
Collapse
|