SwePub
Sök i SwePub databas

  Utökad sökning

Träfflista för sökning "WFRF:(Kriström Berit) srt2:(2015-2019)"

Sökning: WFRF:(Kriström Berit) > (2015-2019)

  • Resultat 1-25 av 26
Sortera/gruppera träfflistan
   
NumreringReferensOmslagsbildHitta
1.
  •  
2.
  • Albertsson-Wikland, Kerstin, et al. (författare)
  • Mortality Is Not Increased in Recombinant Human Growth Hormone-treated Patients When Adjusting for Birth Characteristics
  • 2016
  • Ingår i: Journal of Clinical Endocrinology and Metabolism. - : ENDOCRINE SOC. - 0021-972X .- 1945-7197. ; 101:5, s. 2149-2159
  • Tidskriftsartikel (refereegranskat)abstract
    • Objective: This study aimed to investigate whether reported high mortality in childhood recombinant human GH (rhGH)-treated patients was related to birth-characteristics and/or rhGH treatment. Design and Setting: We sought to develop a mortality model of the Swedish general population born between 1973 and 2010, using continuous-hazard functions adjusting for birth characteristics, sex, age intervals, and calendar year to estimate standardized mortality ratio (SMR) and to apply this model to assess expected deaths in Swedish rhGH-treated patients with idiopathic isolated GH deficiency (IGHD), idiopathic short stature (155) or born small for gestational age (SGA). Participants:The general population: Swedish Medical Birth Register (1973-2010: 1 880 668 males; 1 781 131 females) and Cause of Death Register (1985-2010). Intervention Population: Three thousand eight hundred forty-seven patients starting rhGH treatment between 1985 and 2010 and followed in the National GH Register and/or in rhGH trials diagnosed with IGHD (n = 1890), ISS (n = 975), or SGA (n=982). Main Outcome Measures: Death. Results: Using conventional models adjusting for age, sex, and calendar-year, the SMR was 1.43 (95% confidence interval, 0.89-2.19), P = .14, observed/expected deaths 21/14.68. The rhGH population differed (P amp;lt; .001) from the general population regarding birth weight, birth length, and congenital malformations. Application of an Advanced Model: When applying the developed mortality model of the general population, the ratio of observed/expected deaths in rhGH-treated patients was 21/21.99; SMR = 0.955 (0.591-1.456)P = .95. Model Comparison: Expected number of deaths were 14.68 (14.35-14.96) using the conventional model, and 21.99 (21.24-22.81) using the advanced model, P amp;lt; .001, which had at all ages a higher gradient of risk per SD of the model, 24% (range, 18-42%; P amp;lt; .001). Conclusions: Compared with the general Swedish population, the ratio of observed/expected deaths (21/21.99) was not increased in childhood rhGH-treated IGHD, ISS, and SGA patients when applying an advanced sex-specific mortality model adjusting for birth characteristics.
  •  
3.
  • Albertsson-Wikland, Kerstin, 1947, et al. (författare)
  • Mortality is not increased in rhGH-treated patients when adjusting for birth characteristics.
  • 2016
  • Ingår i: The Journal of clinical endocrinology and metabolism. - : The Endocrine Society. - 1945-7197 .- 0021-972X. ; 101:5, s. 2149-2159
  • Tidskriftsartikel (refereegranskat)abstract
    • Objective: This study aimed to investigate whether reported high mortality in childhood recombinant human GH (rhGH)-treated patients was related to birth-characteristics and/or rhGH treatment. Design and Setting: We sought to develop a mortality model of the Swedish general population born between 1973 and 2010, using continuous-hazard functions adjusting for birth characteristics, sex, age intervals, and calendar year to estimate standardized mortality ratio (SMR) and to apply this model to assess expected deaths in Swedish rhGH-treated patients with idiopathic isolated GH deficiency (IGHD), idiopathic short stature (155) or born small for gestational age (SGA). Participants:The general population: Swedish Medical Birth Register (1973-2010: 1 880 668 males; 1 781 131 females) and Cause of Death Register (1985-2010). Intervention Population: Three thousand eight hundred forty-seven patients starting rhGH treatment between 1985 and 2010 and followed in the National GH Register and/or in rhGH trials diagnosed with IGHD (n = 1890), ISS (n = 975), or SGA (n=982). Main Outcome Measures: Death. Results: Using conventional models adjusting for age, sex, and calendar-year, the SMR was 1.43 (95% confidence interval, 0.89-2.19), P = .14, observed/expected deaths 21/14.68. The rhGH population differed (P < .001) from the general population regarding birth weight, birth length, and congenital malformations. Application of an Advanced Model: When applying the developed mortality model of the general population, the ratio of observed/expected deaths in rhGH-treated patients was 21/21.99; SMR = 0.955 (0.591-1.456)P = .95. Model Comparison: Expected number of deaths were 14.68 (14.35-14.96) using the conventional model, and 21.99 (21.24-22.81) using the advanced model, P < .001, which had at all ages a higher gradient of risk per SD of the model, 24% (range, 18-42%; P < .001). Conclusions: Compared with the general Swedish population, the ratio of observed/expected deaths (21/21.99) was not increased in childhood rhGH-treated IGHD, ISS, and SGA patients when applying an advanced sex-specific mortality model adjusting for birth characteristics.
  •  
4.
  • Andersson, Björn, 1939, et al. (författare)
  • Seasonal variations in vitamin D in relation to growth in short prepubertal children before and during first year growth hormone treatment
  • 2015
  • Ingår i: Journal of Endocrinological Investigation. - : Springer Science and Business Media LLC. - 0391-4097 .- 1720-8386. ; 38:12, s. 1309-1317
  • Tidskriftsartikel (refereegranskat)abstract
    • Purpose This study investigated the relationship between seasonal variations in 25-hydroxyvitamin D (25(OH) D) levels and growth in prepubertal children during both the pretreatment year and the first year of GH treatment. Methods The study included 249 short prepubertal children with a broad range of GH secretion, GH(max) during a 24 h profile median 23; range 1-127 mU/L, 191 boys (mean age +/- SD, 8.6 +/- 2.6 years), 58 girls (7.5 +/- 1.9 years) receiving GH treatment (mean 43 mu g/kg/day; range 17-99 mu g/kg/day). Serum 25(OH) D was measured using an automated IDS-iSYS immunoassay. Results 25(OH) D levels showed seasonal variation, and decreased significantly during GH treatment. 25(OH) D levels at start and first year reduction in 25(OH) D, correlated (-) with the first year growth response during treatment. The degree of GH secretion capacity within our study population of mainly non-GH deficient children and 25(OH) D sufficient (67 +/- 29 nmol/L) had no influence on 25(OH) D levels. Growth during GH treatment were independent of seasonal variations in 25(OH) D. Multiple regression analysis showed that 25(OH) D levels at treatment start, together with auxological data and IGF-binding protein-3(SDS), explained 61 % of the variation in first year gain in height(SDS). Conclusion 25(OH) D levels were associated with first year growth response to GH and may be a useful contribution to future growth prediction models.
  •  
5.
  • Ankarberg-Lindgren, Carina, et al. (författare)
  • Estradiol matrix patches for pubertal induction : stability of cut pieces at different temperatures
  • 2019
  • Ingår i: Endocrine Connections. - : Bioscientifica. - 2049-3614. ; 8:4, s. 360-366
  • Tidskriftsartikel (refereegranskat)abstract
    • Objective: Transdermal estradiol patches are primarily designed for adult women. No low-dose patches are licensed for pubertal induction in hypogonadal girls. Low doses can be achieved by cutting a matrix patch into smaller pieces. However, the manufacturers do not guarantee stability or utility of cut estradiol patches. The aim of the study was to assess 1-month stability of cut estradiol patches from four different manufacturers in the laboratory at room temperature (+21 degrees C) and at an elevated temperature (+35 degrees C).Design and methods: Estraderm MX 50 mu g, Systen 50 mu g and Oesclim 25 mu g matrix patches were cut into eight pieces while Estradot 50 mu g small patches were cut in half. The cut patches were stored in their respective pouches at +21 degrees C or at +35 degrees C for up to 1 month. The estradiol drug was extracted from the patch by ethyl acetate n-hexane and determined by radioimmunoassay.Results: Storage at +21 degrees C or +35 degrees C up to 1 month did not reduce the estradiol concentration in Estraderm MX, Systen and Oesclim patches. However, although the estradiol in Estradot patches was not affected by storage at +21 degrees C, at +35 degrees C, estradiol decreased by 57% (+/- 1%) in cut pieces.Conclusions: Unused Estraderm MX, Systen and Oesclim patch pieces may be stored for at least 1 month at <=+35 degrees C. Where estradiol patches for children are not available, cut pieces of these or similar patches can be used for pubertal induction. The Estradot patch was too small to properly cut into low doses and not stable in elevated temperatures.
  •  
6.
  •  
7.
  • Berglund, Staffan K., et al. (författare)
  • Marginally low birth weight increases the risk of underweight and short stature at three and a half years of age
  • 2016
  • Ingår i: Acta Paediatrica. - : John Wiley & Sons. - 0803-5253 .- 1651-2227. ; 105:6, s. 610-617
  • Tidskriftsartikel (refereegranskat)abstract
    • AIM: Little is known about the long-term health of marginally low birth weight (LBW) children. This study characterised growth among infants weighing 2,000g-2,500g and explored the prevalence and predictors of sustained growth restriction.METHOD: This prospective observational trial followed the weight and height of 281 Swedish marginally LBW children from birth to 3.5 years of age. Children with a standard deviation score (SDS) for body mass index or height below -2 were considered underweight and short respectively.RESULTS: The mean SDS for weight and height showed a rapid increase before 12-19 weeks of age. The most rapid weight gain was in infants born small for gestational age. However, at 3.5 years of age, 9.5% of the children remained underweight and 6.5% had short stature. Regression models showed that slow weight gain before 19 weeks of age was the strongest predictor for lasting underweight, while slow height gain before 19 weeks of age and male sex were associated with short stature.CONCLUSION: Marginally LBW infants were more likely to be underweight and have a short stature at 3.5 years of age and the absence of catch-up growth during the first five months after birth identified those at highest risk.
  •  
8.
  • Borrás Pérez, Maria Victoria, et al. (författare)
  • Ten years of clinical experience with biosimilar human growth hormone : a review of safety data
  • 2017
  • Ingår i: Drug Design, Development and Therapy. - : Dove Press Ltd. - 1177-8881. ; 11, s. 1497-1503
  • Forskningsöversikt (refereegranskat)abstract
    • Safety concerns for recombinant human growth hormone (rhGH) treatments include impact on cancer risk, impact on glucose homeostasis, and the formation of antibodies to endogenous/exogenous GH. Omnitrope (R) (biosimilar rhGH) was approved by the European Medicines Agency in 2006, with approval granted on the basis of comparable quality, safety, and efficacy to the reference medicine (Genotropin (R)). Additional concerns that may exist in relation to biosimilar rhGH include safety in indications granted on the basis of extrapolation and the impact of changing to biosimilar rhGH from other rhGH treatments. A substantial data set is available to fully understand the safety profile of biosimilar rhGH, which includes data from its clinical development studies and 10 years of post-approval experience. As of June 2016, 106,941,419 patient days (292,790 patient-years) experience has been gathered for biosimilar rhGH. Based on the available data, there have been no unexpected or unique adverse events related to biosimilar rhGH treatment. There is no increased risk of cancer, adverse glucose homeostasis, or immunogenic response with biosimilar rhGH compared with the reference medicine and other rhGH products. The immunogenicity of biosimilar rhGH is also similar to that of the reference and other rhGH products. Physicians should be reassured that rhGH products have a good safety record when used for approved indications and at recommended doses, and that the safety profile of biosimilar rhGH is in keeping with that of other rhGH products.
  •  
9.
  • Chaplin, John, 1955, et al. (författare)
  • Growth Hormone Treatment Improves Cognitive Function in Short Children with Growth Hormone Deficiency
  • 2015
  • Ingår i: Hormone Research in Paediatrics. - : S. Karger AG. - 1663-2818 .- 1663-2826. ; 83:6, s. 390-399
  • Tidskriftsartikel (refereegranskat)abstract
    • Background/Aims: We investigated the association between cognition and growth hormone (GH) status and GH treatment in short prepubertal children with broadly ranging GH secretion. Methods: A total of 99 children (age 3-11 years), 41 with GH deficiency (GHD) and 58 with idiopathic short stature (ISS), were randomized to a fixed dose (43 mu g/kg/day) or a prediction model-guided individualized dose (17-100 mu g/kg/day) and followed up for 24 months. In a longitudinal and mixed within-and between-subjects study, we examined clinical effect size changes, measured by Cohen's d, in full-scale IQ (FSIQ) and secondary IQ indices. Results: Significant increases giving medium effect size in FSIQ (p = 0.001, Cohen's d = 0.63), performance IQ (p = 0.001, Cohen's d = 0.65) and processing speed (p = 0.005, Cohen's d = 0.71) were found in the GH-deficient group. In contrast, perceptual organization only increased in the ISS group (p = 0.001, Cohen's d = 0.53). Baseline IQ was normally distributed with small but significant differences between the groups: GH-deficient children had lower FSIQ (p = 0.042) and lower performance IQ (p = 0.021). Using multiple regression analysis, 40% of the variance in delta processing speed scores (0-24 months) was explained by GH(max) and IGF-I-SDS at baseline. Conclusion: IQ, specifically fluid intelligence, increased in the GH-deficient children. The pretreatment status of the GH/IGF-I axis was significantly predictive for these changes. (C) 2015 S. Karger AG, Basel
  •  
10.
  • Collett-Solberg, Paulo F., et al. (författare)
  • Diagnosis, Genetics, and Therapy of Short Stature in Children : A Growth Hormone Research Society International Perspective
  • 2019
  • Ingår i: Hormone Research in Paediatrics. - : S. Karger. - 1663-2818 .- 1663-2826. ; 92:1, s. 1-14
  • Tidskriftsartikel (refereegranskat)abstract
    • The Growth Hormone Research Society (GRS) convened a Workshop in March 2019 to evaluate the diagnosis and therapy of short stature in children. Forty-six international experts participated at the invitation of GRS including clinicians, basic scientists, and representatives from regulatory agencies and the pharmaceutical industry. Following plenary presentations addressing the current diagnosis and therapy of short stature in children, breakout groups discussed questions produced in advance by the planning committee and reconvened to share the group reports. A writing team assembled one document that was subsequently discussed and revised by participants. Participants from regulatory agencies and pharmaceutical companies were not part of the writing process. Short stature is the most common reason for referral to the pediatric endocrinologist. History, physical examination, and auxology remain the most important methods for understanding the reasons for the short stature. While some long-standing topics of controversy continue to generate debate, including in whom, and how, to perform and interpret growth hormone stimulation tests, new research areas are changing the clinical landscape, such as the genetics of short stature, selection of patients for genetic testing, and interpretation of genetic tests in the clinical setting. What dose of growth hormone to start, how to adjust the dose, and how to identify and manage a suboptimal response are still topics to debate. Additional areas that are expected to transform the growth field include the development of long-acting growth hormone preparations and other new therapeutics and diagnostics that may increase adult height or aid in the diagnosis of growth hormone deficiency.
  •  
11.
  • Dalin, Frida, 1984-, et al. (författare)
  • Clinical and immunological characteristics of Autoimmune Addison's disease : a nationwide Swedish multicenter study
  • 2017
  • Ingår i: Journal of Clinical Endocrinology and Metabolism. - : Oxford University Press. - 0021-972X .- 1945-7197. ; 102:2, s. 379-389
  • Tidskriftsartikel (refereegranskat)abstract
    • CONTEXT: Studies on clinical and immunological features of Autoimmune Addison's disease (AAD) are needed to understand the disease burden and increased mortality.OBJECTIVE: To provide upgraded data on autoimmune comorbidities, replacement therapy, autoantibody profiles and cardiovascular risk factors.DESIGN, SETTING AND PARTICIPANTS: Cross sectional, population-based study. 660 AAD patients were included utilizing the Swedish Addison Registry (SAR) 2008-2014. When analyzing cardiovascular risk factors, 3,594 individuals from the population-based survey in Northern Sweden, MONICA (MONItoring of Trends and Determinants of CArdiovascular Disease), served as controls.MAIN OUTCOME MEASURE: Prevalence of autoimmune comorbidities and cardiovascular risk factors. Autoantibodies against 13 autoantigens were determined.RESULTS: Sixty percent of the SAR cohort consisted of females. Mean age at diagnosis was significantly higher for females than for males (36.8 vs. 31.1 years). The proportion of 21-hydroxylase autoantibody positive patients was 83% and 62% of patients had one or more associated autoimmune diseases, more frequently coexisting in females (p<0.0001). AAD patients had lower BMI (p<0.0001) and prevalence of hypertension (p=0.027) compared with controls. Conventional hydrocortisone tablets were used by 89% of patients; with the mean dose 28.1±8.5 mg/day. The mean hydrocortisone equivalent dose normalized to body surface was 14.8±4.4 mg/m(2)/day. Higher hydrocortisone equivalent dose was associated with higher incidence of hypertension (p=0.046).CONCLUSIONS: Careful monitoring of AAD patients is warranted to detect associated autoimmune diseases. Contemporary Swedish AAD patients do not have increased prevalence of overweight, hypertension, T2DM or hyperlipidemia. However, high glucocorticoid replacement doses may be a risk factor for hypertension.
  •  
12.
  • Decker, Ralph, 1968, et al. (författare)
  • Early increase of the bone formation marker PINP is in a higher degree related to growth response compared to bone mineralization in GH treated prepubertal children
  • 2015
  • Ingår i: Horme Research in Paediatrics. - : S. Karger AG. - 1663-2818 .- 1663-2826. ; 84:Suppl 1
  • Konferensbidrag (övrigt vetenskapligt/konstnärligt)abstract
    • Background: It has been reported that short-term increases of the bone formation markers intact amino-terminal propeptide of type I procollagen (PINP), bone-specific alkaline phosphatase (BALP) and osteocalcin display different temporal patterns. In adults, the biphasic model of GH action in bone remodelling shows that GH treatment results initially in an increased bone resorption with a concomitant bone loss, which later on is followed by increased bone formation. In children, little is known how bone remodelling takes place. Objective and hypotheses: Bone formation markers reflect different events during osteogenesis, and respond with different time courses during anabolic GH treatment. Method: The study population comprised 128 short prepubertal children (age range 3−11 years; 90 boys, 38 girls) who participated in a longitudinal, prospective, multicenter study in individual GH dosing1, TRN 98-0198-003. The investigated children had either normal or reduced levels of GH secretion. Data from the first 2 years of GH treatment were analyzed. The bone markers were measured using the IDS-iSYS automatic system (Immunodiagnostic Systems)2. The DXA derived variable bone mineral density (BMD) was measured by Lunar DPX-L or Lunar Prodigy. Results: The bone markers PINP, BALP, osteocalcin and 25-hydroxyvitamin D (25(OH)D) at start and deltaPINP at 3 months of GH treatment explained 63% of the growth response at 2 years (p<0.0001), while only 26% of the variation in BMD response after 2 years of treatment was explained (p<0.0001). Conclusion: Bone markers at start of GH treatment, and the 3 months increase of PINP were associated with both growth response and bone mineralization after 2 years of treatment, but with different magnitude of impact on these anabolic GH effects.
  •  
13.
  • Decker, Ralph, 1968, et al. (författare)
  • GH dose reduction maintains normal prepubertal height velocity after initial catch up growth in short children.
  • 2019
  • Ingår i: The Journal of clinical endocrinology and metabolism. - : The Endocrine Society. - 1945-7197 .- 0021-972X. ; 104:3, s. 835-844
  • Tidskriftsartikel (refereegranskat)abstract
    • GH responsiveness guides GH dosing during the catch-up growth (CUG) period; however, little is known regarding GH dosing during the prepubertal maintenance treatment period.To evaluate if standard deviation score (SDS) channel parallel growth with normal height velocity can be maintained following CUG by reducing GH dose by 50% in children receiving doses individualized based on estimated GH-responsiveness during the catch-up period.and settings: Prepubertal children (n=98; 72 boys) receiving GH during CUG (GH-deficient (n=33); non-GH-deficient (n=65)), were randomized after 2-3 years to either a 50% reduced individualized (GHRID; n=27; 20 boys) or unchanged individualized dose (GHUID; n=38; 27 boys). Another 33 children (25 boys) continued on a standard weight-based dose, 43 µg/kg/day (GHFIX).The primary endpoint was the proportion of children with ΔheightSDS within ±0.3 at 1 year after GH-dose reduction, versus two control groups: GHUID and GHFIX. The hypothesis was that heightSDS could be maintained within ±0.3 with a reduced individualized GH dose.For the intention-to-treat population at 1 year, 85% of the GHRIDgroup maintained ΔheightSDS within ±0.3 versus 41% in the GHUIDgroup, p=0.0055 and 48% in the GHFIXgroup, p=0.0047. ΔIGF-ISDS in the GHRIDgroup was (mean±SD) -0.75±1.0 at 3 months, p=0.003 and at 1 year -0.72±1.2, compared to the GHUIDgroup 0.15±1.2, p=0.005, and for the GHFIXgroup 0.05±1.0, p=0.02.Channel parallel growth, i.e. normal height velocity, and IGFSDS levels within ±2 were maintained after completed CUG using a 50% lower individualized dose than used during the CUG period.
  •  
14.
  • Donaldson, M., et al. (författare)
  • Optimal Pubertal Induction in Girls with Turner Syndrome Using Either Oral or Transdermal Estradiol: A Proposed Modern Strategy
  • 2019
  • Ingår i: Hormone Research in Paediatrics. - : S. Karger AG. - 1663-2818 .- 1663-2826. ; 91:3, s. 153-163
  • Tidskriftsartikel (refereegranskat)abstract
    • Background: Most girls with Turner syndrome (TS) require pubertal induction with estrogen, followed by long term replacement. However, no adequately powered prospective studies comparing transdermal with oral 17 beta-estradiol administration exist. This reflects the difficulty of securing funding to study a rare condition with relatively low morbidity/mortality when competing against conditions such as cancer and vascular disease. Protocol Consensus: The TS Working Group of the European Society for Paediatric Endocrinology (ESPE) has agreed to both a 3-year oral and a 3-year transdermal regimen for pubertal induction. Prerequisites include suitable 17 beta-estradiol tablets and matrix patches to allow the delivery of incremental doses based on body weight. Study Proposal: An international prospective cohort study with single centre analysis is proposed in which clinicians and families are invited to choose either of the agreed regimens, usually starting at 11 years. We hypothesise that pubertal induction with transdermal estradiol will result in better outcomes for some key parameters. The primary outcome measure chosen is height gain during the induction period. Analysis: Assessment of the demographics and drop-out rates of patients choosing either oral or transdermal preparations; and appropriate analysis of outcomes including pubertal height gain, final height, liver enzyme and lipid profile, adherence/acceptability, cardiovascular health, including systolic and diastolic blood pressure and aortic root diameter and bone health. Conclusion: The proposed model of prospective data collection according to internationally agreed protocols aims to break the current impasse in obtaining evidence-based management for TS and could be applied to other rare paediatric endocrine conditions. (C) 2019 S. Karger AG, Basel
  •  
15.
  • Duchén, Karel, et al. (författare)
  • Using a spontaneous profile rather than stimulation test makes the KIGS idiopathic growth hormone deficiency model more accessible for clinicians
  • 2017
  • Ingår i: Acta Paediatrica. - : John Wiley & Sons. - 0803-5253 .- 1651-2227. ; 106:9, s. 1481-1486
  • Tidskriftsartikel (refereegranskat)abstract
    • Aim: Children treated with a growth hormone (GH) for idiopathic growth hormone deficiency (IGHD) may be monitored with the first-year prediction model from the Pfizer International Growth Database (KIGS) using auxology, age, GH dose and the maximum GH concentration from a stimulation test (GH(max)stim). We tested the hypothesis that using a 12-hour spontaneous profile (GH(max)12h) would be as accurate. Methods: We studied 98 prepubertal Swedish children (78boys) aged2-12 years enrolled in KIGS. The first-year growth was predicted using the GH(max) from the GHprofile and a stimulation test, and both of these were compared separately with the observed growth response. Results: The increased height observed in the first year was 0.74 standard deviation scores (SDS), and the studentised residuals for the predicted and observed growth with GH(max)stim (-0.16 SDS) and GH(max)12h (-0.22) were similar. Individual predictions calculated with stimulated or spontaneous GH(max) showed a significant correlation (r = 0.80). Conclusion: We validated the KIGS IGHD prediction model and found that the stimulated GH(max) peak can be reliably replaced by the GH(max) 12h with similar accuracy. This makes the model more accessible for clinicians, who can then provide realistic expectations for the growth response during the first year of treatment.
  •  
16.
  •  
17.
  •  
18.
  • Kriström, Berit, et al. (författare)
  • Growth hormone deficiency in children
  • 2018. - 2
  • Ingår i: Encyclopedia of Endocrine Diseases. - : Elsevier. - 9780128122006 - 9780128121993 ; , s. 66-80
  • Bokkapitel (refereegranskat)abstract
    • Growth hormone (GH) is a highly dynamic hormone, influencing growth and metabolism. GH secretion varies with biological maturity and nutritional state, illness, stress, and physical activity. GH deficiency (GHD) may be observed in the neonatal period or later throughout life. Thus, knowledge on normal physiology is necessary. Before any GH investigation, other conditions interfering with GH secretion or action must be ruled out. GH stimulates liver secretion of IGF-I and IGFBP-3; their serum concentrations may give information on the GH/IGF-I axis. GH investigation can be performed by stimulation tests or by estimating spontaneous GH secretion. If GHD is diagnosed, substitution therapy is possible. However, dosing is complicated by individual responsiveness to both GH and IGF-I, and possibly also varies with the growth phases. Auxological and biochemical variables associated with GH responsiveness have been retrieved from large cohorts of GH-treated children. There are models for prediction of growth response, indirectly informing us on individual responsiveness that can guide GH dosing from the start of treatment. More than 30 years of GH treatment have shown satisfactory safety, although the age of treated subjects and time for follow-up make continued follow-up studies necessary.
  •  
19.
  •  
20.
  •  
21.
  • Lundberg, Elena, et al. (författare)
  • Growth hormone (GH) dose-dependent IGF-I response relates to pubertal height gain
  • 2015
  • Ingår i: Bmc Endocrine Disorders. - : Springer Science and Business Media LLC. - 1472-6823. ; 15
  • Tidskriftsartikel (refereegranskat)abstract
    • Background: Responsiveness to GH treatment can be estimated by both growth and Delta IGF-I. The primary aim of the present study was to investigate if mimicking the physiological increase during puberty in GH secretion, by using a higher GH dose could lead to pubertal IGFs in short children with low GH secretion. The secondary aim was to explore the relationship between IGF-I, IGFBP-3 and the IGF-I/IGFBP-3 ratio and gain in height. Methods: A multicentre, randomized, clinical trial (TRN88-177) in 104 children (90 boys), who had received GH 33 mu g/kg/day during at least 1 prepubertal year. They were followed from GH start to adult height (mean, 7.5 years; range, 4.6-10.7). At onset of puberty, children were randomized into three groups, to receive 67 mu g/kg/day (GH(67)) given once (GH(67x1); n = 30) or divided into two daily injection (GH(33x2); n = 36), or to remain on a single 33 mu g/kg/ day dose (GH(33x1); n = 38). The outcome measures were change and obtained mean on-treatment IGF-I-SDS, IGFBP3(SDS) and IGF-I/IGFBP3 ratio(SDS) during prepuberty and puberty. These variables were assessed in relation to prepubertal, pubertal and total gain in height(SDS). Results: Mean prepubertal increases 1 year after GH start were: 2.1 IGF-I-SDS, 0.6 IGFBP3(SDS) and 1.5 IGF-I/IGFBP3ratio(SDS). A significant positive correlation was found between prepubertal Delta IGFs and both prepubertal and total gain in height(SDS). During puberty changes in IGFs were GH dose-dependent: mean pubertal level of IGF-I-SDS was higher in GH67 vs GH(33) (p = 0.031). First year pubertal Delta IGF-I-SDS was significantly higher in the GH(67) vs GH33 group (0.5 vs -0.1, respectively, p = 0.007), as well as Delta IGF-I-SDS to the pubertal mean level (0.2 vs -0.2, p = 0.028). In multivariate analyses, the prepubertal increase in 'Delta IGF-I-SDS from GH start' and the 'GH dose-dependent pubertal Delta IGF-I-SDS' were the most important variables for explaining variation in prepubertal (21 %), pubertal (26 %) and total (28 %) gain in height(SDS). Conclusion: The dose-dependent change in IGFs was related to a dose-dependent pubertal gain in height(SDS). The attempt to mimic normal physiology by giving a higher GH dose during puberty was associated with both an increase in IGF-I and a dose-dependent gain in height(SDS).
  •  
22.
  • Lundberg, Elena, 1961- (författare)
  • Growth hormone responsiveness in children : results from Swedish multicenter clinical trials of growth hormone treatment
  • 2017
  • Doktorsavhandling (övrigt vetenskapligt/konstnärligt)abstract
    • The general aims of the thesis were to study GH responsiveness by estimation of pharmacokinetics and bioavailability of injected recombinant human GH (rhGH), of growth response as gain in heightSDS during childhood and puberty, and IGF-I response as change in circulating IGF-ISDS and IGFBP3SDS. Methods Short children were recruited during 1988–1999 into two national randomized multicentre clinical trials on growth until adult height. A group of 117 GHD patients who had been treated from prepuberty with a single GH dose of 33μg/kg/day for at least 1 year were randomized at onset of puberty either to remain on this dose regimen or to an increased dose, GH67μg/kg/day, administered once daily or divided into two doses, GH33x2μg/kg/day. Data on IGF-ISDS and IGF binding protein 3 (IGFBP3)SDS were available from 111 patients and analysed as stated below. The 151 short prepubertal non-GHD patients were randomized into three groups: untreated controls, GH33 or GH67μg/kg/day. A subpopulation from both trials, 128 patients examined annually in Gothenburg, formed the study sample on GH uptake. They received sc GH injections to obtain 16–24 hour GH curves and the GH pharmacokinetics and bioavailability was calculated. Results: A dose-dependent effect on Cmax was found with great intra- and inter-individual variability. Of the Cmax variability, 43% was explained by the rhGH dose and proxies for injection depth. Median bioavailability of the injected dose was 71%, with great variation, mainly dependent on injection depth. In the IGHD group a dose-dependent difference in pubertal gain in heightSDS was found, with mean of 0.8 for the GH67 group and 0.4 for GH33, p<0.01. The mean total gain in heightSDS during treatment was 1.9 for GH67 and 1.4 for GH33, p<0.01. A dose-dependent pubertal ΔIGF-ISDS was 0.5 vs −0.1, p=0.007, correlating to pubertal gain in heightSDS, p=0.003; and was the most important variable to explain the variation in pubertal gain in heightSDS. In the non-GHD group the ΔIGF-ISDS from baseline to mean study level was dose-dependent 2.07 vs 1.20, p=0.001; and correlated negatively with baseline values of IGF-ISDS, rho= -0.56 for GH67, p=0.001, vs rho= -0.82 for GH33, p=0.0001, and correlated positively with gain in heightSDS in both GH-treated groups, rho= 0.42, p<0.001. In multivariable regression analyses, ΔIGF-ISDS was always an important explanatory variable for long-term growth response from the prepubertal period until adult height, while the IGF-ISDS study level per se was not. Conclusion: Growth response to GH treatment was dose dependent with great variability between patients. More pubertal growth was attained by an increased rhGH dose, mimicking the physiology of healthy children, in whom GH secretion rate increases during puberty. This resulted in a gain in IGF-ISDS closely correlating to pubertal gain in heightSDS in both IGHD and non-GHD patients. A broad range in GH responsiveness was found for both growth and IGF response in both diagnostic groups, but lower in the non-GHD group. Higher uptake of a given GH dose was observed after a deep injection and a higher GH concentration. These results are clinically applicable for individuals who remain short close to onset of puberty; by identifying and deeply injecting a rhGH dose that accounts for individual responsiveness, we can stimulate an increment in IGF-ISDS that correlates to gain in heightSDS during puberty.
  •  
23.
  •  
24.
  •  
25.
  • Norjavaara, Ensio, 1954, et al. (författare)
  • Sex Steroid Replacement Therapy in Female Hypogonadism from Childhood to Young Adulthood.
  • 2016
  • Ingår i: Endocrine development. - : S. Karger AG. - 1662-2979 .- 1421-7082. ; 29, s. 198-213
  • Tidskriftsartikel (refereegranskat)abstract
    • The overall goal of pubertal sex hormone replacement therapy (HRT) in girls is not only about development of secondary sexual characteristics, but also to establish an adult endocrine and metabolic milieu, as well as adult cognitive function. Estradiol (E2) is the first choice for HRT compared to ethinyl estradiol (EE2). E2 is the most potent endogenous estrogen in the circulation, with established levels during spontaneous puberty. Transdermal E2, compared to oral administration, is the first choice to start pubertal HRT. Transdermal application avoids liver exposure to supraphysiologic estrogen concentrations and provides a more physiologic mechanism for hormone delivery. By cutting E2 matrix patches in doses of 0.05-0.07 µg/kg or administrate E2 gel in doses of 0.1 mg/day, serum concentrations of E2 seen in early spontaneous puberty can be obtained. Patches can be removed in the morning and thereby mimic the normal circadian rhythm. For those clinics with access to sensitive E2 determinations methods (extraction followed by radioimmunoassay or mass spectrometry) monitoring the attained E2 serum levels is recommended in order to optimally mimic the levels seen in early puberty as well as growth velocity, breast and uterus development. Mid- and late pubertal HRT is obtained by increased doses of E2, adding cyclic oral or transdermal progestin, as well as testosterone gel over the pubic area if indicated.
  •  
Skapa referenser, mejla, bekava och länka
  • Resultat 1-25 av 26
Typ av publikation
tidskriftsartikel (22)
konferensbidrag (1)
doktorsavhandling (1)
forskningsöversikt (1)
bokkapitel (1)
Typ av innehåll
refereegranskat (15)
övrigt vetenskapligt/konstnärligt (11)
Författare/redaktör
Kriström, Berit (23)
Albertsson-Wikland, ... (7)
Albertsson-Wikland, ... (6)
Lundberg, Elena, 196 ... (4)
Bang, Peter (3)
Gustafsson, Jan (3)
visa fler...
Mårtensson, Anton (3)
Dahlgren, Jovanna, 1 ... (3)
Norgren, Svante (3)
Pehrsson, Nils-Gunna ... (3)
Lundberg, Elena (3)
Duchén, Karel (2)
Odén, Anders, 1942 (2)
Jonsson, Björn (2)
Gustafsson, Jan, 194 ... (2)
Höybye, Charlotte (2)
Niklasson, Aimon, 19 ... (2)
Sävendahl, Lars (2)
Kriström, Berit, 194 ... (2)
Ankarberg-Lindgren, ... (2)
Olcén, Per (1)
Kämpe, Olle (1)
Domellöf, Magnus (1)
Hofman, Paul L. (1)
Janson, Annika (1)
Landin-Olsson, Mona (1)
Rönnelid, Johan (1)
Johannsson, Gudmundu ... (1)
Winqvist, Ola (1)
Hallgren, Åsa (1)
Bensing, Sophie (1)
Tuvemo, Torsten (1)
Juul, Anders (1)
Dahlqvist, Per (1)
Gelander, Lars, 1956 (1)
Westphal, Otto, 1935 (1)
Åman, Jan (1)
Albertsson Wikland, ... (1)
Savendahl, Lars (1)
Niklasson, Aimon (1)
Dahlgren, Jovanna (1)
Oden, Anders (1)
Norjavaara, Ensio, 1 ... (1)
Ekwall, Olov, 1968 (1)
Söderberg, Stefan (1)
Chaplin, John, 1955 (1)
Isaksson, Magnus (1)
Norman, Mikael (1)
Bidlingmaier, Martin (1)
Cohen, Pinchas (1)
visa färre...
Lärosäte
Umeå universitet (24)
Göteborgs universitet (10)
Uppsala universitet (6)
Karolinska Institutet (5)
Linköpings universitet (4)
Chalmers tekniska högskola (2)
visa fler...
Örebro universitet (1)
Lunds universitet (1)
visa färre...
Språk
Engelska (26)
Forskningsämne (UKÄ/SCB)
Medicin och hälsovetenskap (26)

År

Kungliga biblioteket hanterar dina personuppgifter i enlighet med EU:s dataskyddsförordning (2018), GDPR. Läs mer om hur det funkar här.
Så här hanterar KB dina uppgifter vid användning av denna tjänst.

 
pil uppåt Stäng

Kopiera och spara länken för att återkomma till aktuell vy