Annals of Medicine

ISSN: 0785-3890 (Print) 1365-2060 (Online) Journal homepage: http://www.tandfonline.com/loi/iann20

Evaluation of Growth Hormone Secretion and Treatment Päivi Tapanainen & Mikael Knip To cite this article: Päivi Tapanainen & Mikael Knip (1992) Evaluation of Growth Hormone Secretion and Treatment, Annals of Medicine, 24:4, 237-247, DOI: 10.3109/07853899209149951 To link to this article: http://dx.doi.org/10.3109/07853899209149951

Published online: 08 Jul 2009.

Submit your article to this journal

Article views: 8

View related articles

Full Terms & Conditions of access and use can be found at http://www.tandfonline.com/action/journalInformation?journalCode=iann20 Download by: [Laurentian University]

Date: 17 April 2016, At: 08:35

Review Article

Evaluation of Growth Hormone Secretion and Treatment

Downloaded by [Laurentian University] at 08:35 17 April 2016

Paiivi Tapanainen and Mikael Knip

The secretion of growth hormone (GH) is regulated by a complex system that includes both neurotransmitters and feedback by hormonal and metabolic substrates. Over the last few years it has been recognized that GH release varies over a wide spectrum from deficient to excessive secretion. The diagnosis of GH deficiency is based on a combination of anthropometric and clinical signs on the one hand and an inadequate stimulated and/or spontaneous GH secretion on the other. There is no distinct boundary between deficient and sufficient GH secretion. The cut-off limit for normal GH release is accordingly relative and has increased over the past decade from 5 to 10 ,ugh. The effect of GH therapy on growth can be evaluated only after treatment for at least 6 months. There is, therefore, an indisputable need for methods that would retiect growth response soon after the start of treatment. There are several promising biochemical candidates, e.g. the aminoterminal propeptide of type Ill procollagen, the carboxyterminal propeptide of procollagen I and the bone Gla-protein, which may turn out to be useful early indicators of the growth response to long-term GH therapy. Key words: growth hormone; secretion; regulation; therapy; response; prediction. (Annals of Medicine 2 4 237447,1992)

Introduction The increased availability of human growth hormone (hGH) produced by recombinant DNA techniques has raised new questions about the indications for its use. Classically, GH deficiency has been considered the only indication. New insights into the regutation and physiology of GH secretion have shown that there is no distinct boundary between deficient and sufficient GH secretion (1) and accordingly the definition of GH deficiency is a relative and not an absolute one. It has also been argued that the indications for GH therapy should be based on GH responsiveness and the extent of handicap associated with short stature rather than on abnormalities in GH secretion (2). This approach may, however, result in extensive use of GH on heafthy short children. In addition, a GH-deficient child may have a better growth response to therapy than a short child with normal amounts of GH. So assessment of GH secretion remains important in the diagnosis of growth disorders, especially for the evaluation of the extent of GH deficiency. GH therapy is expensive and tedious and its effect on growth can be evaluated only after a minimum of six From the Department of Pediatrics, University of Oulu, SF90220 Oulu, Finland. Address and reprint requests: Mikael Knip, M.D., Department of Pediatrics, University of Oulu, SF-90220 Oulu, Finland.

months’ use. In order to reduce costs and spare those children for whom such treatment is unnecessary, methods are needed that would reflect the growth response soon after treatment starts, especially if it is to be used as a crucial criterion for long-term treatment.

Regulation of GH Secretion GH secretion is regulated by a complex neuroendocrine control system that includes both neurotransmitters and feedback by hormonal and metabolic substrates [Fig. I ). The final common pathway for the integration of these signals involves two neuropeptides, which are hypophysiotropic hormones. Growth hormone-releasing hormone (GHRH), a 40-44 residue peptide, has a stimulating effect, while somatostatin, a tetradecapeptide, inhibits GH secretion. GHRH GHRH was originally characterized as a 44-amino acid peptide that was isolated, together with several other biologically active fragments (GHAHI -40-OH and GHRH1-37-NH2), from an islet cell tumour of the pancreas in an acromegalic patient without a pituitary adenoma (3). A 40-amino acid fragment was also isolated

Tapanainen Knip

238

Neurotransmitters, glucose, amino acids, lipids, sleep, temperature J i V

1 1 1 1 1 Hypothalamus

Pituitary

Peripheral tissues

I+

CeIl differentiation

Downloaded by [Laurentian University] at 08:35 17 April 2016

Clunaf expansion

Figure 1, The hypothalamo-pituitary-IGF-l-axis.A complex neuroendocrine system comprising bofh neurotransmitters and feedback by hormonal and metabolic substrates regulates

growth hormone secretion from the anterior pituitary. The hypothalamic signals are integrated into a common pathway involving two neuropeptides; the stimulatory growth horrnonereleasing hormone and the inhibitory somatostatin. In peripheral tissues growth hormone has both direct and indirect effecls of which the tatter are mediated by insulin-likegrowth factor I.

Somatostatin Somatostatin, a hypothalamic factor with an inhibiting action on pituitary GH secretion, was isolated from ovine hypothalamus (23) and found to be a tetradecapeptide (S-14) with a cyclic structure and an intramolecular disulphide bond between its two cysteine residues. Brazeau et al. were the first to report the effects of somatostatin on GH release using cultured pituitary cells (23); since then it has been shown on several occasions that somatostatin has a physiological role, acting via the hypophyseal portal system to regulate GH secretion from the anterior pituitary gland. The responses of GH secretion to all physiological and pharmacological stimuli are known to be inhibited by somatostatin. Somatostatin blocks the GH responses to exercise, arginine, insulininduced hypoglycaemia (IIH) and GHRH in man (24-27), and immunoreactive somatostatin has also been measured in peripheral plasma (28, 29). Liapi et al. demonstrated a negative correlation between peripheral plasma somatostatin levels and GH response to GHRH stimulation in children (281,but Rosskamp et al. found no correlation between increments in plasma somatostatin and GHRH in response to a mixed meal (29).

interactions Between GHRH and Somatostatin

and characterized from another turnour recognized under similar circumstances (4). The primary structure of human hypothalmic GHRH was subsequently shown to be identical to that of the 44-amino acid peptide originally isolated from the pancreatic tumour (5).The biological activity of the 40 and 44-amino acid peptides seems to be indistinguishable when a variety of in vitro systems are used (6-8)and when injected into humans (9-1 1). In humans and other primates, most perikarya containing GHRH are located in the mediobasal hypothalamus (12), although GHRH has also been identified in extra-CNS loci. Cow GHRH immunoreactivity have been reported in several segments of the gastrointestinal tract (13, 14). Quantification of GHRH requires sensitive, specific radioimmunoassays. Several laboratories have published data on immunoreactive GHRH levels in plasma (13-1 8), but only a few reports include validation by high pressure liquid chromatography (HPLC) (16, 17).Accordingly, it has not been verified in all studies whether the measured amounts are really those of GHRH or whether they represent GHRH metabolites or non-specific imrnunoreactivity. It has been shown recently in rats that mechanical ablation of GHRH neurons in the medial basal hypothalamus results in a 70% reduction in circulating ir-GHRH levels (1s), showing that most peripheral imrnunoreactivity could originate from hypothalmic sources. Some authors have claimed that changes in circulating ir-GHRH concentrations correlate with the secretion of GH (20-22),suggesting that peripheral plasma ir-GHRH measurements do reflect hypothalmic function.

The contributions of GHRH and somatostatin to the generation of GH secretory pulses have been examined in several species. They have been most intensively studied in the rat, however, with the aid of two techniques that selectively exclude the effect of one of the hormones, passive immunization and hypothalmic destruction (30).It has been shown by means of antisomatostatin serum injections that pulsatile GH secretion is preserved despite an increase in basal GH levels (31), whereas administration of anti-GHRH serum has little effect on already low basal GH levels but inhibits secretory bursts (25).Continuous infusion of a large dose of GHRH to unanaesthetized rats pretreated with anti-somatostatin serum has been shown to increase the concentration of GH in plasma (32). These findings have been interpreted as showing that somatostatin is importan1 for maintaining low basal levels of GH but that pulsatile GH secretion is dependent on GHRH. Lesions of the medial preoptic area of the hypothalamus lead to a depletion in median eminence somatostatin, a rise in GH levels for some days and abolition of the expected pattern of pulsatile GH secretion (33). These findings are best explained by bihormonal control of GH secretion with asynchronous periodic release of GHRH and somatostatin. Pulses of GH secretion would accordingly be expected at times of maximal GHRH secretion and minimal somatostatin secretion (30). Analysis of the various components of GH feedback regulation and of the contribution of GHRH and somatostatin to pulsatile GH secretion cannot be accomplished as easily in primates as in rodents, though mounting evidence suggests that the mechanisms are similar (30). A bolus injection of GHRH in man produces an acute dose-dependent secretory response (34), while administration of GHRH as a constant infusion produces a persistent effect on GH secretion characterized by

GH Secretion and Treatment enhanced pulsatile secretion and a rise in baseline GH secretion, in contrast to the al'most undetectable baseline GH levels normally observed (35, 36). Our own results suggest that the secretion of ir-GHRH is pulsatile and that GHRH seems to affect nocturnal pulsatile GH secretion in growing children (22).

Downloaded by [Laurentian University] at 08:35 17 April 2016

Role of Various Neurotransmitter Systems in the Regulation of GH Secretion In addition to specific neurohormones, a series of brain neurotransmitters are involved in the neural control of GH secretion (37). These have both stimulatory and inhibitory effects on GH release, exerted mainly via GHRH and/or somatostatin. Alternatively, neurotransmitters may act directly at the pituitary level (37). Catecholamines and acetylcholine seem to be major factors in the neuroregulation of GH secretion in humans (see 38), and it has been shown with regard to the catecholaminergic system that clonidine, an alpha2receptor agonist, induces a clear-cut rise in plasma GH concentration both in animals and in man (see 38).Clonidine induces GH release in children, but this does not seem to be mediated by GHRH (15). Our own results also suggest that other factors mediate the effect of clonidine (18). Oral L-Dopa administration has been found to result in moderate increases in the amounts of plasma ir-GHRH and GH (15, 18); thus it is likely that L-Dopa stimulated GHRH secretion, resulting in GH release.

239

specific receptors in the tissues (46). The gene for hGH is found on the long arm of chromosome 17 (47).

GH Secretion in Humans GH is secreted in pulses at all stages of life, being secreted nocturnally in episodes occurring within 90-120 min of the onset of sleep and associated with slow wave sleep (48). Pulses of considerable magnitude which are unrelated to sleep, activity, stress or nutrient intake nevertheless occur throughout the day. Pulsatile GH secretion is variable, though patterns tend to be relatively constant in individual subjects (49). Women secrete more GH than men, and older men and women secrete less GH than their young counterparts (17, 50). Reports on the effect of puberty on spontaneous GH secretion are conflicting. One group has concluded that puberty has no effect on GH secretion (51),whereas several other investigations, including our own, have found increased 24-h or overnight GH concentrations during puberty in both boys and girls (22, 52-54). The increase in GH secretion at puberty seems to represent an amplitude-modulated phenomenon that is relatively independent of changes in pulse frequency (53). Such changes may be secondary to the action of sex steroid hormones which modulate the responsiveness of the somatotroph to endogenous GHRH, the amount of GHRH secreted, or the inhibitory tone of somatostatin (50). It has also been shown that pubertal children have higher basal plasma ir-GHRH concentration than prepubertal children (22,55).

Insulin-like Growth Factors Insulin-like growth factors (IGF-I and IGF-ll) are small polypeptides structurally related to insulin. IGFs exert insulin-like, differentiative and mitogenic actions on their target tissues (39). They are produced in the liver and many other tissues, and are partially dependent on GH (39). Many tissues and cell lines possess IGF receptors, designated as either type I or type 11, through which IGFs exert their biological actions (40). A recently recognized class of proteins which have high affinity and specificity for IGFs, known as IGF-binding proteins (ICFBPs), have been shown to be involved in the modulation of the proliferative and mitogenic effects of IGFs on cells (41, 42). The major factor regulating the postnatal production of IGF-I is GH, which enhances IGF-I synthesis in various organs and tissues, in primary cell cultures and in established cell lines (see 43). Amounts of IGF-I in the blood are raised in acromegaly and depressed in patients with GH deficiency (44). IGF-I is also currently thought to be the main mediator of the growth-promoting actions of GH (45).

GH Secretion and Somatic Growth The GH Molecule The human GH protein consists of a single chain of 191 amino acid residues with a total molecular weight of approximately 22,000. It circulates in the blood partly as a complex with a binding protein, and it binds to its

Effectsof GH GH has both direct and indirect effects. Its characteristic metabolic effects are anabolic, lipolytic and glucose sparing. It has also been shown to stimulate longitudinal bone growth directly by increasing the differentiation of epiphyseal growth plate precursor cells and indirectly by increasing the responsiveness of chondrocytes to IGF-I and enhancing local production of the IGF-I that stimulates the clonal expansion of differentiating chondrocytes (45). GH also has rapid and direct stimulatory effects on amino acid transport and protein synthesis which may reflect its growth-promoting'property (56).Many of the growth-promoting actions of GH are indirect, being mediated by IGFs. Both IGF-I and IGF-II can stimulate growth in hypophysectomized animals, although the effect of IGF-II is marginal (57, 58).

Assessment of GH Secretion Before 1963 the diagnosis of GH deficiency was based entirely on clinical findings. Since then, the development of radioimmunoassays for GH has led to the establishment of laboratory criteria for this diagnosis (see 59). GH secretion can be evaluated by means of pharmacological stimulation tests and physiological tests. Endogenous 24-h or overnight GH secretion has recently been used to evaluate spontaneous GH secretion.

240

Tapanainen Knip

Pharmacological Stimulation Tests

Downloaded by [Laurentian University] at 08:35 17 April 2016

The finding that hypoglycaemiawas a potent stimulus for the secretion of GH (60) rapidly led to the application of this technique to the diagnosis of GH deficiency in children. Many other pharmacological stimuli have also been used, such as clonidine (61), levodopa (L-Dopa) (62), glucagon (63), arginine (64) and dinoprostone (prostaglandinE2)(65). The mechanisms by which these tests exert their effect are not always well defined (15, 18) and some tests may also be hazardous for the subjects studied (66).Pharmacological tests may give conflicting results, and for this reason it has been recommended that more than one test of GH secretion should be performed (67).

the differential diagnosis of hypothalmic vs. pituitary GH deficiency (78,79). Ranke et al. have concluded that testing with GHRH offers a safe and convenient way of diagnosing GH deficiency in clinically suspected cases (80), while others are of the opinion that it is not a very helpful test in this respect (81, 82). Tests performed on small groups of children with GH deficiency have shown that the response to a single dose of GHRH is heterogeneous, possibly due to the varied aetiologies of GH deficiency and to the duration and extent of GH deficiency (83). Schriock et al. have claimed that it is not possible to conclude whether GH deficiency is of hypothalamic or pituitary origin on the basis of a single GHRH test, since in some cases the somatotrophs can regain their function after priming with GHRH (84).

Physiological Tests

Measurement of IGF-I Levels

The stimuli usually used for assessing physiological secretion are sleep (68) and exercise (69).It has been shown that electroencephalographic(EEG) monitoring of sleep improves the efficiency of this physiological test (70). Hyperthermia in the form of a hot water bath, has also been used previously as a means of GH stimulation in children (71). We have shown that GH release in this relatively.mild,non-invasive test appears to be mediated by GHRH and conclude that plasma GHRH measurements in the presence of hypertherrnia could be used as a test to identify possible hypothalamic defects in the regulation of GH secretion (18). Endogenous 24-h or overnight GH secretion has been used to evaluate spontaneous GH release (72-74). A recent report points to a relationship between total 24-h GH secretion and the growth of normal children over a wide range of growth rates (74), although Shulman and Bercu found no correlation between height and 24-h .GH secretion (75). It has also been suggested that measurements of 24-h GH secretion may identify a subgroup of GH-deficient children who are not detected by provocative testing, and who may yet respond to exogenous GH therapy with improved linear growth (73). On the other hand, a recent report indicates that the measurement of spontaneous OH release offers no diagnostic advantage over stimulation tests (59). Measurements of urinary GH have increased in popularity recently (76),mainly because immunoassays for the measurement of GH in urine have improved markedly with the availability of more sensitive methods. GH is secreted in a pulsatile manner, however, and the amplitude of the peaks and troughs is important for growth. These data are lost with the use of urinary GH measurements. It has been suggested that the measurement of urinary OH could be a useful, non-invasive screening test for the evaluation of GH secretion (76). On the other hand, there are reports that show that further methodological improvements' are required to allow reliable evaluation of GH secretory abnormalities through the use of nocturnal urinary GH excrection (77).

The widespread availability of sensitive, specific radioimmunoassays for IGF-I (85) and IGF-II (86) has resulted in numerous investigations into the regulation of their serum concentrations by GH and other factors. It is clear that serum IGF-I concentrations are regulated by GH and that they may reflect endogenous GH secretlon (87). The diagnostic usefulness of these measurements is limited, however, since their concentration is also affected by other factors. IGF-I concentration is age-dependent in man and relatively low in young children, which makes interpretationof the results difficult in this age group (88). IGF-I levels also depend on other hormones and, primarily, on the nutritional state (89). Serum IGF-II concentrations are much less GH-dependent than those of IGF-I, and are reduced in only about half of all patients with GH deficiency. The combination of IGF-I and IGF-II measurements has been reported to permit better discrimination between normal, normal short and GHdeficient children (90).The measurement of IGF-I is helpful as a screening test for GH deficiency in all age groups, but because of its lack of specificity, low values must be interpreted with caution. In contrast, the routine measurement of serum IGF-II concentration is of limited use in the clinical evaluation of GH secretion (90).

The GHRH Test GHRH has been used widely as a stimulation test for GH secretion in adults and children in recent years, and for

Measurement of insulin-like Growth Factor Binding Protein 3 (IGFBP-3) A specific radioimmunoassay has been developed for IGFBP-3 and used to investigate its diagnostic potential for the evaluation of growth disorders (91). Serum levels of IGFBP-3 are dependent on age and nutrition, and are evidently also related to GH concentrations, in that they appear to reflect integrated GH secretion over several days (91). They are low in patients with GH deficiency, but children with normal variant short stature have higher amounts. Patients with familial tall stature have concentrations in the upper normal range and patients with acromegaly in the supranormal range. These results have led to the conclusion that serum IGFBP-3 may be a highly informative screening measurement for GH deficiency (92). There seems to be less overlap for circulating IGFBP-3 concentrations than for IGF-I concentrations between GH-deficient children and those with normal GH values.

241

GH Secretion and Treatment

Growth Hormone Deficiency There are very few children who have deletion of the GH gene and produce no GH at all, and most of those previously identified as GH-deficient do produce small amounts in response to physiological stimuli and provocative pharmacological testing (see 93). There is no clear cut-off point, but rather a continuum whkh varies from severe deficiency to mild or partial insufficiency and normal to excessive GH secretion (Fig. 2) (94). Zadik et al. have claimed that some subjects among children with short stature have a normal GH response to pharmacological stimuli but do not spontaneously secrete enough GH for optimal growth

Downloaded by [Laurentian University] at 08:35 17 April 2016

(95). The characteristic clinical features of children with deletion of the GH gene (96) are that they are extremely small even when well nourished and present certain facial traits. Correspondingly, the clinical appearance of children with severe GH deficiency is remarkably uniform. They are short (relative height < - 3 SDs), have varying degrees of bone age retardation, a skinfold thickness at or above the 90th percentile and a characteristic appearance (97). Children with tess severe GH deficiency may look normal. Many children of short stature fail to meet the criteria for GH deficiency. This group includes children with biologically inactive GH, children who have low spontaneous GH secretion, children of normal variant short stature and those of small-for-date shori stature. The assessment of GH secretion has been discussed above. The traditional definition of GH deficiency has been based on a slow growth rate coupled with an inadequate GH response to at least two provocative tests. The diagnostic 'cut-off level for GH in the provocative tests has gradually been increased from the initial 5 pg/I up to 10 pg/l, which is now commonly used

(see 97). The stringent criteria required for the diagnosis of GH deficiency have recently been broadened, mainly because of the increased supply of biasynthetic hGH, and some groups dso believe that the only way to diagnose GH deficiency is a 6-month therapeutic trial with hGH. Assessment of G H secretion staFts with auxological data and a clinical examination, and several measurements of height over a period of 6-12 months are needed to evaluate a child's growth rate. Rates of growth and maturation are important criteria in the differential diagnosis of short stature (see 97). The determination of bone age is also valuable, although bone age can vary widely in subjects who meet the criteria in every other respect and respond to GH therapy and, in a few children, may even be advanced retative to chronological age (see 97).

Growth-stimulating Therapy and Predictors of Response to Therapy GH Treatment The effectiveness of hGH on height growth In a GHdeficient patient was first reported by Raben in 1958 (98). Nationalry organized pituitary collection programmes were subsequently arranged in many countries to provide the large number of pituitary glands essential to meet the increasing demand for extracted human pituitary GH (hpGH). HpGH was withdrawn from clinical use in May 1985 following the diagnosis of CreutzfeldtJakob disease in one patient in the United Kingdom and several in the USA who had been treated with relatively impure early preparations of hpGH (99, 100). Since then, advances in recombinant technology have enabled the production of recombinant sornatotropin with full biological activity and extremely high purity (101). Recombinant

Nnrrnrrl

/ =I Severe GH deficiency Provocative testing

24-h profile

0

11

I I I I

I

1 1 1

I L I I

I

I I t I

Partial GH insufficiency 1 @ o r ? I I I 1 I

I

I I I 1

I

+ I

normal

1 I 1

I

t I

Figure 2. GH release varies over a wide spectrum from deficient to excessive secretion. Severe GH deficiency is characterized by severely decreased spontaneous GH secretion and no response to pharrnacotogical or physiologicalstimulation, while in partial GH insufficiency spontaneous secretion is less reduced and there is some GH response to stimulation (reproduced from reference 94 with the permission of the author and the publisher).

Downloaded by [Laurentian University] at 08:35 17 April 2016

242

Tapanainen

somatotropin has been shown to have good growthpromoting effects and low immunogenicity in several multicentre trials (102-1 04). Since the initial publication on the effectiveness of hGH in promoting growth in stature (99) many reports on treatment trials with hGH have been published (105-1 09). Most of these deal with short-term growth responses, and the number in which patients have been followed up to their final height is still limited. Optimal treatment should be aimed at achieving normal adult stature, but it is apparent from recent data that only about half of patients with GH deficiency have grown to normal height (107-1 09). Before the production of recombinant hGH, growth failure due to GH deficiency was the only universally accepted therapeutic indication for GH treatment. By 1983, reports of fewer than 300 non-GH-deficient children with short stature who had received GH treatment had been published (110). Now many multicentre trials cover short non-GH-deficient children with recombinant GH, although only short-term results are available so far (103, 104, 111-1 15). Assessing the effects of treatment in the reported cases is complicated by, e.g. the short duration of therapy and the lack of a universal definition of a good response to GH therapy. Absolute growth velocities are usually presented rather than relative growth velocities when assessing the response to treatment. Though it is obvious that GH therapy will promote growth in some children with undefined short stature (1.13), we still lack criteria for selecting suitable children for therapy. Short children with low spontaneous GH secretion have been treated with hGH, and AlbertssonWikland reports a positive growth response in 29 out of 31 cases and a negative correlation between growth response and the amount of hGH secreted spontaneously during a 24-h period before treatment (1 14). In contrast, Butenandt (115) observed only a small increase in height velocity over 1 year. Research with large randomized trials is needed to determine the benefit of GH treatment for children without documented GH deficiency. Patients with Turner’s syndrome have also been treated with GH. Earlier attempts to treat girls with the syndrome in this way failed to establish convincingly that it was beneficial (116, 117), but these involved only a few cases treated for a short time under uncontrolled conditions. Recently, increased growth rates have been documented during the treatment of girls with Turner syndrome using hGH (1 18,119).

Other Growth-stimulating Treatment It has been shown on a number of occasions that some patients with organic lesions of the hypothalamo-pituitary axis have specific defects in either the synthesis or the delivery of endogenous GHRH. Furthermore, most children with idiopathic panhypopituitarism or isolated GH deficiency, as diagnosed by the existence of little or no GH response to pharmacological tests, will respond to GHRH analogues. The truncated 29-residue analogue in particular is much smaller than GH and retains the

Knip

normal feedback sensitivity of GH to high concentrations of circulating GH (120). Long-term pulsatile infusion of GHRH (1-40) by means of a portable pump has been shown to increase linear growth velocity in GH-deficient children, with a concomitant rise in circulating IGF-I (121). Reports on an improvement in growth velocity upon subcutaneous administration of GHRH have come from many groups using dose frequencies of once daily (122, 123), twice daily (124) or 3-h pulses, both overnight (124) and for 24-h periods (125). There are still unanswered questions under investigation concerning GHRH therapy, such as the optimal dose, mode of application and the effect of various GWRH analogues. Clonidine treatment for children with short stature is controversial. Subjects with constitutional delay have been treated with orally administrated clonidine and their linear growth rate has been found to increase (126, 127). Pescovitz et al. (128), however, have shown in a doubleblind, placebo-controlled trial that long-term administration of a single daily dose of clonidine does not result in any increase in GH production or in accelerated growth velocity. IGF-I has recently been used to treat some types of short stature. Laron et al. (129), treating nine patients with Laron-type dwarfism, showed that the lack of GH receptors in this condition does not apply to IGF-I receptors or post-receptor pathways, and suggested that long-term treatment with IGF-I may therefore be beneficial in this condition. Walker et al. (130) also found that infusion of IGF-I had an anabolic potential in patients with GH insensitivity and that IGF-I may be effective in promoting linear growth in such children.

Clinical Assessment of the Response to Growthstimulating Treatment Although children with GH deficiency have been treated with substitution therapy for over 25 years, the major predictors of the response to treatment have not been clearly defined. The determination of body height and the calculation of growth rates are still the major parameters for the assessment of the response to GH therapy. To date, these parameters have been derived from serial estimations of body height, usually with a stadiometer, taken several weeks or months apart, the growth rate being described by the difference between two statural heights divided by the respective time interval. This measure of growth rate, also called growth velocity, is assumed to be an adequate expression of growth (131). Response to treatment, however, depends on the child’s pretreatment growth rate and on the amount of GH administered and the frequency of administration (132). There is also evidence that monthly measurements of height do not differ significantly from random figures, and that without a knemometer the minimum time necessary to detect a response to GH therapy is 6 months (133).

Biochemical Predictors of the Response to Growthstimula ting Treatment GH treatment usually results in increases in circulating IGF-I concentrations (134), and the acute IGF-I response

Downloaded by [Laurentian University] at 08:35 17 April 2016

GH Secretion and Treatment

has been found to predict chronic IGF-I responses during GH therapy (135). Initial reports (134jindicated a positive correlation between GH-stimulated IGF-I levels and subsequent growth velocity, but later ones (135-137)suggested that the circulating IGF-I response does not reliably predict the growth response. On the other hand, close correlations were recently found between the relative increases in the amounts of serum IGF-1 and IGF-II and the growth increase during GH treatment in short chjldren without GH deficiency (114) Accordingly, present knowledge indicates that the GHinduced IGF-I response may be a useful. predictor of response to treatment in children with undefined short stature but not in GH-deficient children. A preliminary report suggests, however, that pretreatment serum IGF-I concentrations correlate inversely with growth response to GH in patients with hypopituitarism if the IGF-I concentrations are measured after acid gel chromatography of the samples (138). Somatic growth results from the proliferation of cells and from the deposition af material in the extracellular matrix. This process takes place in both bone and soft tissues. Type 111 collagen is a major collagenous component of the latter and is found together with type I collagen and a number of other constituents. The serum concentrations of the aminoterminal propeptide of type Ill collagen (PIIINP) have been found to be related to growth rate in healthy children (139). We have observed close correlations between the increment in serum PlllNP amounts after 5 weeks and growth velocity after both 6 and 12 months of GH treatment, suggesting that the GH-induced increase in serum PlllNP concentrations may be a potential early indicator of the growth response to treatment (140). Other investigations have also shown that the measurement of serum PIIINP, particularly in children receiving exogenous GH, may provide useful additional biochemical information during growth (141, 142). Trivedi et al. suggest that the most appropriate application of PIllNP could be for monitoring prepubertal children receiving exogenous GH therapy. Serum concentrations of the carboxyterminal propeptide of type I collagen (PICP) have been reported to be abnormally low both in GH-deficient children (143) and in children with retarded growth due to inflammatory bowel disease (144). Our preliminary results show that serum PICP and PlllNP concentrations seem to be of equal value for the prediction of the long-term response to GH therapy (145). Bone Gla-protein (BGP) is a small protein that accounts for approximately 25% of the noncollagenous protein found in adult bone (146). It is synthesized by the osteoblasts and incorporated into the bone matrix A fraction is also released into the circulation, however, where it can be measured by radioimmunoassay. Serum 5GP concentrations change in relation to age and sex, with a pattern that resembles the height velocity curves for children (147). A recent study has shown that the GH-induced increment in serum BGP concentrations at 3 months was significantly related to growth response after 12 months of treatment and suggests that serum BGP values could be a hetpful biochemical marker for predicting the growth response to long-term GH therapy (148). Future research will provide

243

information on which candidate protein may turn out ro be the most useful for predicting response to GH therapy. Such a reliable predictor should enable identification of GH-responsive short children within a few weeks and could also facilitate the estimation of individual optimal doses of exogenous GH. We thank the Sigrid Juselius Foundation for supporting our

studies.

References 1. Hindmarsh P, Smith PH, Brook CGD, et al. The relationship between height velocity, and growth hormone secretion in short prepubertal children. Clin Enndocrrnol 1987;27:581-91. 2. Allen DB, frost NC. Growlh hormone therapy for short stature: panacea or Pandora's box? d Pediafr 1990; 117: 16-21. 3. Guillemln R, Brazeau P, Bohlen P, Esch F, Llng N, Wehrenberg WB. Growlh hormone-releasing factor from a human pancreatic tumor that caused acromegaly. Science 1982; 218: 585-7. 4. Rivler J, Spiess J, Thorner

M,Vale W. Characterization of a growth hormone-releasing factor from a human pancreatic islet tumor. Nature 1982;300: 276-8. 5. Ling N, Esch F, Bijhlen P. Brazeau P, Wehrenberg WP, Gulllernin R. Isolation, primary structure, and synthesis of human hypothalmic sornatocrinin; growth hormonereleasing factor. Proc Natl Acad Sci USA 1984; 81: 4302-6. 6. Brazeau P, Ling N, Bbhlen P, Esch F, Ylng S-Y, Guillemin R. Growth hormone-releasing factor, somatocrinin. releases pituitary growth hormone in vitro. Proc Nad ACad SCi USA 1982;79: 7909-1 3. 1. Spless J, Rivier J, Vale W. Characterization of rat hypothalamic growth hormone-releasing factor. Nature 1983:

303:532-5. Frohman LA, Jansson J - 0 . Growth hormone-releasing hormone. Endomin Rev 1986; 7: 223-52. 9. Thorner MO, Rivier J, Spless J, et al. Human pancreatic

a.

growth honone-releasing factor selectively stimulates growth hormone secretion in man. Lancet 983:1: 24-8. 10. Rooenthat SM. Schribck EA, Kaplan SL, Guillemin R, Grumbsch MM. Synthetic human pancreas growth hormone-releasing factor (hpGRH 1-44 -NH2) stimulates growth hormone secretion in normal men. J CIin Endocrinol Metab 1983; 57: 677-9. 11. Chlhara K, Kadama H, Kaji H, e l el. Augmentation by propranol of growth hormone-releasing hormone- (1 -44)NH 2-induced growth hormone release in normal short and normal children. J Clin Endocrind Metab 1985; 61: 229-33. 12. Blach 8, Galllard RC, Brazeau P,Lln HD. Llng N. Topographical and ontogenetic study of the neurons producing growth hormone-releasing factor in human hypothalamus. Regd Pepr 1 984; 8:21-31. 13. Frohman LA, Thominet JL, Webb CB, st al. Metabolic clearance and plasma disappearance rates of human pancreatic tumor growth hormone-releasing factor in man. JClinlnvest f964;73:1304-11. 14. Thorner YO, Frohman LA, Leong DA, et al. Extrahypolhalamic growth hormone-releasing factor [GBF) secretion is a rare cause of acromegaly: plasma GRF levels in 177 acrornegalic patients. d Clin Endocrind Metab 1984; 59:846-9. 15. Dannadleu M, Evain-Brim D, Tonon MC, Veudry H, Job J-C. Variations of plasma growth hormone (GH)-releasing

Tapanainen Knip

244

factor levels during GH stimulation tests in children. J Clin Endocrinol Metab 1984;69:1 132-4. 16. Penny ES, Patience RL, Sopwith AM, Wass JAH, Besser GM, Rees, LH. Characterization by high performance liquid chromatography of circulating growth hormonereleasing factors in a human plasma. J Endocrinol 1985;

105:Rl-R4. 17. Leppaluoto J, Tapanalnen P, Knip M. Heat exposure elevates plasma immunoreactive growth hormone-releasing hormone levels in man. J Clin Endocrinol Metab 1987;

Downloaded by [Laurentian University] at 08:35 17 April 2016

65:1035-8. 18. Tapanalnen P, Knip M, Lautala P, Leppaluoto J. Variable plasma growth hormone (GH)-releasing hormone and GH response to clonidine, L-dopa, and insulin in normal men. J Clin EndocrinolMetab 1988;67:845-9. 19. Lina L, Carolto MC, Dall'Ara A, Cocchi D. Significanceof circulating immunocreative growth hormone-releasing hormone: CNS vs. peripheral source. 70th Annual Meeting of the Endocrine Society, New Orleans, LA, 8-11 June 1988,abstract 261. 20. Saito H, Saito S, Yamazakl R, Hosol E. Clinical value of radioimmunoassay of plasma growth hormone-releasing factor. Lancet 1984;2:401 -2. 21. Smith PJ, Pringle PJ, Rafferty B, Brook CGD. The relationship between endogenous immunocreative GRH and GH secretion. J Endocrinol 1985;104,Suppl: 86. 22. Tapanainen P, Rantala H, Leppaluoto J, Lautala P, Kaar ML, Knip M. Nocturnal release of immunoreactive growth hormone (GH)-releasing hormone and GH in normal children. Pediatr Res 1989;26:404-9. 23. Brazeau P, Vale W, Burgus R, et al. Hypothalmic polypeptide that inhibits the secretion of immunoreactive pituitary growth hormone. Science 1973;179:77-9. 24. Hall R, Besser GM, Schally AV, et at. Action of growth hormone-release inhibitory hormone in healthy men and in acromegaly. Lancet 1973;2:581 -4. 25. Prange-Hansen AP, Orskov H, Seyer-Hansen K, Lundbaek K. Some actions of growth hormone-release inhibiting factor. Br Med J 1973;3:523-4. 26. Mortimer CH, Tunbrldge WMB, Carr D, et al. Effects of growth hormone-release-inhibitinghormone on circulating glucagon, insulin, and growth hormone in normal, diabetic, acromegalic, and hypopituitary patients. Lancet 1974; 1 :

697-701. 27. Wehrenberg WB, Brazeau P, Luben R, Bahlen P, Guillemin R. Inhibition of the pulsatile secretion of growth hormone by monoclonal antibodies to the hypothalamic growth hormone releasing factor (GRF). Endocrinology

1982;1 1 1:2147-8. 28. Llapl, C, Evain-Brlon D, Argente J, Vaudry H, Donnadieu M. Negative correlation between peripheral plasma somatostatin levels and GH responses to GHRH stimulation tests in children. Acta fndocrinol 1986;113:1-4. 29. Rosskamp R, Becker M, Haverkamp F, et al. Plasma levels of growth hormoneireleasing hormone and somatostatin in response to a mixed meal and during sleep in children. Acta Endocrinol 1987;116:549-54. 30. Frohman LA, Downs TR, Katakaml H, Jansson J-0. The interaction of growth hormone-releasing hormone and somatostatin in the regulation of growth hormone secretion. In: lsaksson 0, Binder C, Hall K, Hokfelt 6,eds. Growth hormone: basic and clinical aspects. Amsterdam: Excerpta Medica International Congress Series, 1987;

748:63-77. 31. Ferland L, Labrle F, Jobin M, Arimura A, Schally AV. Physiological role of somatostatin in the control of growth hormone and thyrotropin secretion. Biochem Biophys Res Commun 1976;68:149-56. 32. Wehrenberg WB, Brazeau P, Ling, N, Textor G, Gulllemin R. Pituitary growth hormone response in rats during a 24-hour infusion of growth hormone-releasing factor. Endocrinology 1984;1 14: 1613-6.

33. Katakaml H, Downs TR, Frohman LA. Inhibitory effect of hypothalamic medial preoptic area somatostatin (SRTIF) on GH-releasing hormone (GRH) in the rat. 68th Annual Meeting of the Endocrine Society, Anaheim, CA, 26-27 June 1986.abstract 65. 34. Vance ML, Borges JLC, Kaiser DL, et al. Human pancreatic tumor growth hormone-releasing factor: dose-response relationships in normal man. J Clin €ndocrinol Metab 1984;58:838-44. 35. Vance ML, Kaiser DL, Evans WS, et al. Evidence for a limited growth hormone (GH)-releasing hormone (GHRH)releasable quantity of GH: effects of 6-hour infusions of GHRH on GH secretion in normal man. J Clin Endocrinol Metab 1985;60:370-5. 36. Webb CB, Vance ML, Thorner MO, et al. Plasma growth hormone responses to constant infusions of human pancreatic growth hormone releasing factor: intermittent secretion or response attenuation. J Clin lnvest 1984;74:

96-103. 37. Camannl F, Ghlgo E, Mazza E, et al. Aspects of neurotransmitter control of GH secretion: basic and clinical studies. In: Advances in growth hormone and growth factor research. Muller E, Cocchi D, Locatelli V, eds. Roma-Milano: Pythagora Press, 1989;263-81. 38. Muller EE. Neural control of somatotropic function. Physiol Rev 1987;67:962-1053. 39.Froesch ER, Schmld C, Schwander J, Zapf J. Actions of insulin-like growth factors. Ann Rev Physiol 1985; 47:

443-67. 40. Casella SJ, Han VK, D'Ercole AJ, Svoboda ME, Van Wyk JJ. Insulin-like growth factor II binding to the type I somatomedin receptor: evidence for two high affinity binding sites. J Biol Chem 1986;261:9268-73. 41. Rosenfeld RG, Lamson G, Pham H, et al. Insulin-like growth factor binding proteins. Rec Progr Horrn Res 1990;

46:99-163. 42. Rutanen EM, Pekonen F. Insulin-like growth factors and their binding proteins. Acta Endocrinol 1990;123:7-13. 43. Lamson G, Gludlce LC, Rosenfeld RG. Insulin-like growth factor binding proteins: structural and molecular relationships. Growth factors 1991;5:19-28. 44. Clemmons DR, van Wyk JJ. Factors controlling blood concentrations of somatomedin C. Clin fndocrinol Metab 1984;13:1 13-43. 45. lsaksson OGP, Llndahl A, Nilsson A, lsgaard J. Mechanism of the stirnulatory effect of growth hormone on longitudinal bone growth. Endocrine Rev 1987; 8:

426-38. 46. Hirt H, Kimelman J, Birnbaum MJ, et al. The human growth hormone gene locus: structure, evolution and allelic variations. DNA 1987;6: 59-70. 47. Baumann G, Stolar MW, Amburn K, et al. A specific growth hormone binding protein in human plasma: initial characterization. J Clin fndocrinol Metab 1986; 62:

134-41. 48. Phllllps JA. Regulation of and defects in expression of growth hormone genes. In: lsaksson 0, Binder C, Hall K,

Hokfelt 6,eds. Amsterdam: Excerpta Medica International Congress Series, 111 1987;748:1 1-27. 49. Honda Y, Takahashl K, Takahashl S, et al. Growth hormone secretion during nocturnal sleep in normal subjects. J Clin EndocrinolMetab 1969;29:20-9. 50. Thorner MO, Rogol AD. Neuroendocrine regulation of growth hormone secretion. In: Underwood LE, ed. Human growth hormone, progress and challenges. New York: Marcel Dekker Inc., 1988:1 13-30. 51. Thompson RG, Rodriguez A, Kowarskl A, Migeon CJ, Blizzard RM. Integrated concentrations of growth hormone correlated with plasma testosterone and bone age in preadolescent and adolescent males. J Clin Endocrinol Metab 1972;35:334-7. 52. Zadlk 2, Chalew SA, McCarter J Jr, Melstas M,

Downloaded by [Laurentian University] at 08:35 17 April 2016

GH Secretion and Treatment Kowarskl AA. The influence of age on the 24-hour integrated concentration of growth hormone. J Clin fndocrinol Metab 1985; 60: 51 3-1 6. 53. Albertsson-Wikland K, Rosberg S. Analyses of 24-hour growth hormone profiles in children: relation to growth. J Clin Endocrinol Metab 1988; 67: 493-500. 54. Minuto F, Barreca A, Ferrini S, Mauocchi G, Del Monte P, Giordano G. Growth hormone secretion in pubertal and adult subjects. Acta Endocrinol 1982; 99: 161-5. 55. Argente J, Evaln-Brion D, Munoz-Villa A, Garnier P, Hernandez M, Donnadieu M. Relationship of plasma growth hormone-releasing hormone levels to pubertal changes. J Clin Endocrinol Metab 1986; 63: 680-2. 56. Kostyo JL. The growth-promoting activity of growth hormone. In: Underwood LE,ed. Human growth hormone, progress and challenges. New York: Marcel Dekker Inc., 1988: 63-73. 57. Schoenle E, Zapf J, Froesch ER. Insulin-like growth factors I and II stimulate growth of hypophysectomized rats. Qiabetologia 1982; 23: 199. 58. Schoenle E, Zapf J, Humbel RE, Froesch ER. Insulin-like growth factor I stimulates growth in hypophysectomized rats. Nature 1982; 296: 252-3. 59. Rose SR, Ross JL, Uriarte M, et al. The advantage of measuring stimulated as compared with spontaneous growth hormone levels in the diagnosis of growth hormone deficiency. N Engl J Med 1988; 31 9: 201 -7. 60. Roth J, Glick SM, Yalow RS, Berson SA. Hypoglycemia: a potent stimulus to secretion of growth hormone. Science 1963; 140: 987-8. 61. The Health Services Human Growth Hormone Committee. Comparison of the intravenous insulin and oral clonidine tolerance tests for growth hormone secretion. Arch Dis Child 1981 ; 56: 852-4. 62. Boyd AE, Lebovitz HE, Pfeiffer JB. Stimulation of human growth hormone secretion by L-dopa. N Engl J Med 1970; 283: 1425-9. 63. Vanderschueren-Lodeweyckx M, Wolter R, Malvaux P, et al. The glucagon stimulation test: effect on plasma growth hormone and on immunocreative insulin, cortisol and glucose in children. J fediatr 1974; 85: 182-7. 64. Merimee TJ, Lillicrap DA, Rablnovitz D. Effect of arginine on serum levels of human growth hormone. Lancet 1965;2: 668-70. 65. Hamilton W, Hussein DM. Growth hormone response to prostaglandin E 2. Acta Pediatr Scand 1979; 68: 251 -6. 66. Frasier SD. A review of growth hormone stimulation tests in children. Pediatrics 1974;53: 929-37. 67. Brook CGD, Hindmarsh PC, Smith PJ, Stanhope R. Clinical features and investigation of growth hormone deficiency. Clin Endocrinol Metab 1986; 15: 479-93. 68. Takahashi Y, Kipnis DM, Daughaday' WH. Growth hormone secretion during sleep. J Clin lnvest 1968; 47: 2079-90. 69. Lacey KA, Hewison A, Parkin JM. Exercise as a screening test for growth hormone deficiency in children. Arch Dis Child 1973; 48: 508-1 2. 70. Hindmarsh PC, Smith PJ, Taylor BJ, et al. Comparison between a physiological and a pharmacological stimulus of growth hormone secretion: response to stage IV sleep and insulin-induced hypoglycaemia. Lancer 1985; 2: 1033-5. 71. Geisen K, Meder R. The effect of heat on the level of human growth hormone in the serum. Z Kinderheilkd 1969; 106: 308-1 3. 72. Dacou-Voutetakis C, Drakopoulou M, Maniati M. Growth hormone levels during sleep in children. Pediatr Adolesc Endocrinol 1983; 12: 44-52. 73. Bercu BE, Shulman D, Root AW, Spiliots BE. Growth hormone (GH) provocative testing frequently does not reflect endogenous GH secretion. J Clin Endocrinol Metab 1986;63: 709-1 6.

245

74. Albertsson-Wikland K, Westphal 0, Westgren U. Daily subcutaneous administration of human growth hormone in growth hormone deficient children. Acta Paediatr Scand 1986; 75: 89-97. 75. Schulman DI, Bercu BB. Evaluation of growth hormone secretion: provocative testing vs. endogenous 24-hour growth hormone profile. Acra faediatr Scand 1987; Suppl. 337: 61 -71. 76. Sukegawa I, Hizuka N, Takano K, et al. Urinary growth hormone (GH) measurements are useful for evaluating endogenous GH secretion. J Clin Endocrinol Metab 1988; 66: 1 1 19-23. 77. Tanaka T, Yoshizawa A, Mikl Y, It0 J. Clinical usefulness of urinary growth hormone measurement in short children. Acta faediatr Scand 1990; 366 (Suppl): 155-8. 78. Schriock EA, Hulse JA, Harris DA, Kaplan SL, Grumbach MM. Evaluation of hypothalamic dysfunction in growth hormone (GH)-deficient patients using single versus multiple doses of GH-releasing hormone (GHRH44) and evidence for diurnal variation in somatotroph responsiveness to GHRH in GH-deficient patients. J Clin Endocrinol Metab 1987; 65: 1177-82. 79. Kajiwara S, lgarashi N, lmura E, Sato T. Correlation between pituitary growth hormone reserve and degree of growth failure in children with short stature. Eur J fediatr 1988; 147: 584-87. 80. Ranke MB, Gruhler M, Rosskamp R, et al. Testing with growth hormone-releasing factor (GRH 1-29) NH2.) and somatomedin C measurements for the evaluation of growth hormone deficiency. Eur J fediatr 1986; 145: 485-92. 81. Chatelain P. European collaborative study on the effect of synthetic 1-44 growth-hormone-releasing factor (GRF) on plasma growth hormone (GH) in prepubertal children with growth failure. Pediatr Res 1985; 19: 61 0. 82. Borges JLC, Blizzard RM, Gelato MC, et al. Effects of human pancreatic tumour growth hormone releasing factor on growth hormone and somatomedin C levels in patients with idiopathic growth hormone deficiency. Lancet 1983; 1: 1 19-24. 83. Schriock EA, Lustig RH, Rosenthal SM, Kaplan SL, Grumbach MM. Effect of growth hormone (GH)-releasing hormone (GRH) on plasma GH in relation to magnitude and duration of GH deficiency in 26 children and adults with isolated GH deficiency or multiple pituitary hormone deficiencies: evidence for hypothalamic GRH deficiency. J Clin Endocrinol Metab 1984; 58: 1043-49. 84. Schriock EA, Rosenthal SM, Egli CA, et al. Studies with growth hormone-releasing factor (GRF) in the human: effect of a single pulse, or multiple pulses of GRF on growth hormone (GH) release in normal and GH-deficient children and adults. In: Raiti S, Tolman RA, eds. Human growth hormone. New York: Plenum Publishing Corp., 1986: 387-403. 85. Furlanetto RW, Underwood L, Van Wyk JJ, D'Ercole AJ. Estimation of somatomedin-C levels in normals and patients with pituitary disease by radioimmunoassay. J Clin lnvest 1977; 60: 648-657. 86. Zapf J, Walter H, Froesch ER. Radioimmunological determination of insulin-like growth factors I and II in normal subjects and in patients with growth disorders and extrapancreatic tumor hypoglycemia. J Clin lnvest 1981; 68: 1321 -1 330. 87. Furlanetto RW. Insulin-like growth factor measurements in the evaluation of growth hormone secretion. Horm Res 1990; 33: SUPPI4 , 2 5 3 0 . 88. Furlanetto RW, Marlno JM. Radioimmunoassays of somatomedin C/insulin-like growth factor 1. Methods Enzymoll985; 146: 21 6. 89. Merimee TJ, Zapf J, Froesch ER. Insulin-like growth factors in the fed and fasted states. J Clin Endocrinol Metab 1982; 55: 999-1 002.

Downloaded by [Laurentian University] at 08:35 17 April 2016

246

Tapanainen

90. Rosenfeld RG, Wilson GM, Lee POCHIntz RL. Insulinlike-growth factors I and I1 in evaluation of growth retardation. J Pediatr 1986: 109: 428-33. 91. Blum WF, Ranke MB, Kletzmann K, Ganggel E, Zeisel HJ, Bierlch JR. A specific radioimmunoassay for the growth hormone (GH)-dependent somatomedin-binding protein: it's use for diagnosis of GH deficiency. J Clin EndocrinolMetab 1990; 70: 1292-8. 92. Blum WF, Ranke MB. Use of insulin-like growth factorbinding protein 3 for evaluation of growth disorders. Horm Res 1990; 33, Suppl4: 31-7. 93. Brook CGD, Hindmarsh PC, Smlth PJ. Is growth hormone deficiency a useful diagnosis? Acta Paediatr Scand 1987; Suppl33: 70-5. 94. Vanderschuerer-Lodeweyck 2. The establishment of growth hormone deficiency in children. lnternational Growth Monitor 1990; 1: 3-9. 95. Zadlk Z, Chalew SA, Raitl S, Kowarskl AA. Do short children secrete insufficient growth hormone? Pediatrics 1985; 76: 355-60. 96. llllg R, Prader A, Fernandez A, Zachmann M. Hereditary prenatal growth hormone deficiency with increased growth hormone antibody formation. Symposium der Deutschen Gesellschaft fur Endokrinologie 1970; 16: 246-7. 97. Hindmarsh PC, Brook CGD. Auxological and biochemical assessment of short stature. Acta faediarr Scand 1988; SUPPI343: 73-6. 98. Raben MS. Treatment of a pituitary dwarf with human growth hormone. J Clin Endocrinol Metab 1958; 18: 901-3. 99. Powell-Jackson, Weller J, Kennedy P, Preece MA, Whltcombe EM, Newson-Davis J. Creutzfeldt-Jakob disease after administration of human growth hormone. lancet 1985; 2: 244-6. 100 Gibbs CJ Jr, Joy A, Heffner R, Franko M, et al. Clinical and pathological features and laboratory confirmation of Creutzfeldt-Jakob disease in a recipient of pituitaryderived human growth hormone. N Engl J Med 1985; 313: 734-8. 101. Fryklund L. Production of authentic recombinant somatropin. Acta Paediatr Scand 1987; Suppl331: 5-8. 102. Kapian SL, Underwood LE, August GP, et al. Clinical studies with recombinant-DNA-derived methionyl human growth hormone in growth hormone-deficient children. Lancet 1986; 1: 697-700. 103. Hindmarsh PC, Brook CGD. Effect of growth hormone on short normal children. Br Med J 1987; 295: 573-7. 104. Albertsson-Wlkland K, Aronson S, Nllsson KO, et al. Recombinant somatotropin in treatment of growth hormone deficient children in Sweden and Finland. Acta Paediatr Scand 1980; Suppl347: 176-80. 105. Prader A, tachmann M, Poley JR, llllg R, Sreky J. Longterm treatment with human growth hormone (Raben) in small doses. Evaluation of 18 hypopituitary patients. Helv Paediatr Acta 1967; 22: 423-40. 106. Mllner RDG, Russell-Fraeer T, Brook CGD, et al. Experience with human growth hormone in Great Britain: the report of the MRC working party. Clin Endocrinol1979; 11: 15-38. 107. Ranke MB, Blerich JR. Treatment of growth hormone deficiency. Clin Endocrinol Metab 1986; 15: 495-510. 108. Zupplnger K, Joss E, Aoten A, Schwa- HP, Aebl C. Endresultate bei hypophyzaren Kleinwuchs nach Wachstumshormontherapie. Med Praxis 1980; 34: 1197-204. 109. Lenko HL, Lelstl S, Perheentupa J. The efficiency of growth hormone in different types of growth failure. An analysis of 101 cases. Eur J fediatr 1982; 138: 241-9. 110. Ad HOCCommittee on Growth Hormone Usage, The Lawson Wllklns Pediatric Endocrine Society, and Committee on Drugs. Growth hormone in the treatment of

Knip

short stature. Pediatrics 1983; 72: 891-4. 111. Bierlch JR. Multicentre clinical trial of authentic recombinant somatropin in growth hormone deficiency. Acta Paediatr Scand 1987; Suppl337: 135-40. 112. Van W e t 0, Styne DM, Kaplan SL, Grumbach MM. Growth hormone treatment for short stature. N Engl J Med 1983; 309: 1016-22. 113. Genentech Collaborative Study Group. Idiopathic short stature: results of nine 1-year controlled study of human growth hormone treatment. J Pediatr 1989; 115: 713-9. 114. Albertsson-Wlkland K, Hall K. Growth hormone treatment in short children: relationship between growth and serum insulin-like growth factor I and I1 levels. J Clin Endocrinol Metab 1987; 65: 671-8. 115. Butenandt 0. Die konstitutionelle entwicklungsverzligerung - untersuchungen zur therapie mit wachstumshormon, Klin fediatr 1984; 196: 69-72. 116. Tanner JM, Whltehouse RH, Hughes PCR, Vlnce FP. Effect of human growth hormone treatment for 1 to 7 years on growth of 100 children, with hormone deficiency, low birth-weight, inherited smallness, Turner's syndrome, and other complaints. Arch Dis Child 1971; 46: 745-82. 117. Rudman Q, Goldsmith M,Kutner M, Blackston D.Effect of growth hormone and oxandrolone singly and together on growth rate in girls with X chromosome abnormalities. J Pediatr 1980; 96: 132-5. 118. Raitl S, Moore WV, Van Vllet G, Kaplan SL and The National Hormone and Pitultaty Program. Growthstimulating effects of human growth hormone therapy in patients with Turner's syndrome. J Pediatr 1986; 109: 944-9. 119. Rosenteld RG, Hlntz RL, Johanson AJ, et al. Three year resuits of a randomized prospective trial of methionyl human growth hormone and oxandrolone in Turner syndrome. J Pediatr 1988; 113: 393-400. 120. Ross RJM, Grossman A, Smlth 8, et al. Evidence that growth hormone inhibits the GH response to growth hormone-releasingfactor in man. J. Endocrinol 1985; 107, Suppl 28. 121. Thorner MO, Reschke J, Chltwood J, et al. Acceleration of growth in two children treated with human growth hormone-releasingfactor. N Engl d Med 1985; 312: 4-9. 122. Rochlccloll PE, Tsuber MT, Coude FX, et al. Results of 1-year growth hormone (GH)-releasing hormone (1-44) treatment on growth, somatomedin-C, and 24-hour GH secretion in six children with partial GH-deficiency. J C/in Endocrinol Metab 1987; 65: 268-74. 123. Ross RJ, Rodda C, Tsagarakls S, et al. Treatment of growth hormone deficiency with growth hormone-releasing hormone. Lancet 1987; 1: 5-8. 124. Smith PJ, Brook CG, Rlvier J, Vale W, Thorner MO. Nocturnal pulsatile growth hormone releasing hormone treatment in growth hormone deficiency. Clin Endocrinol 1986; 25: 35-44. 125. Low LCK, Wang C, Cheung PT, et al. Long term pulsatile growth hormone (GH)-releasing hormone therapy in children with GH deficiency. d Clin Endocrinol Metab 1988; 66;611-7. 126. Plntor C, Cella SG, Loche S, et 81. Clonidine treatment for short stature:Lencet 1987; 1; 1226-30. 127. Loche S, Pugglonl R, Fannl T, Cella SG, Muller EE, Plntor C. Augmentation of growth hormone secretion in children with constitutional growth delay by short term clonidine administration: a pulse amplitude-modulated phenomenon. J Clin EndocrinolMetab 1989; 68: 426-30. 128. Pescovltz H,Tan E. Lack of benefit of clonidine treatment for short stature using double-blind, placebo-controlled trial. Lancet 1988; 2: 874-7. 129. Laron Z, Klinger B, Erster B, Anln S. Effect of acute administration of insulin-like growth factor I in patients with Laron-type dwarfism, Lancet 1988; 2: I170-2. 130. Walker JL, Glnalska-Mallnovoka M, Romer TE,

Downloaded by [Laurentian University] at 08:35 17 April 2016

GH Secretion and Treatment Pucilowska JB, Underwood LE. Effects of the infusion of insulin-iike growth factor I in a child with growth hormone insensitivity syndrome (Laron dwarfism). N Engl J Med 1991; 324: 1483-8. 131, Hermanussen M, Geiger-Benoit K, Burmeister J, Sippell WG. Can the knemometer shorten the time for growth rate assessment? Acta Paediatr Scand 1987; SUPPI337:30-6. 132. Albertsson-Wikland K, Isaksson 0, Rosberg S, Westgren U, Westphal 0. Frequency and mode of growth hormone administration: relationship to growth in prepubertal children. Acra Endocrinol 1983: Suppl 256: 21 5. 133. Hermanussen M, Slppell WG, Valk IM. Knemometric monitoring of early effects of human growth hormone on leg length in children with growth hormone deficiency. Lancet 1985;1069-71. 134. D’Ercole Ad, Underwood LE, Van Wyk JJ. Serum somatomedin-C in hypopituitarism and in other disorders of growth. J Pediatr 1977;9 0 375-81 135. Rosenfeld RG, Kemp SF, Hlntz RL. Constancy of somatomedin response to growth hormone treatment of hypopituitary dwarfism, and lack of correlation with growth rate. J Clin Endocrinol Metab 1981 ; 53: 61 1-1 7. 136. Blethen Sl Daughaday WH, Weldon W. Kinetics of the somatomedin Chsulin-like growth factor I: Response to exogenous growth hormone (GH) in GH-deficient children. J Clin Endocrinol Metab 1982;54:986-90. 137. Lodeweyckx M, Wolter R. Effect of growth hormonereleasing factor on plasma growth hormone, prolactin and somatomedin C in hypopituitary and short normal children. Horm Res 1985;22:32-45. 138. Hlntz R, Frane J, Sherman B, and The Genentech Collaborative Group. Pretreatment IGF-I correlates with growth response to GH in hypopituitarism: the importance of acid separation. Pediafr Res 1988;24:522. 139. Riotell J, Nleml S, Trlvedl P, Maentauota 0, Mowat AP, Risteli L. Rapid equilibrium radioimrnunoassay for the amino-terminal propeptide of human type 111 procollagen. Clin Chem 1988;34:71 5-8. ~

247

140. Tapanainen P, Rlsteli L, Knip M, Kl3r M-L, Ristell J. Serum aminoterminal propeptide of type 111 procollagen: a potential predictor of the response to growth hormone therapy. J Clin Endocrinol Metab 1988;67:1244-9. 141. Trivedi P, Hindmarsh P, Risteli J, Risteli L, Mowat A, Brook C. Growth velocity, growth hormone therapy and serum concentrations of the aminoterminal propeptide of type 111 procollagen. J Pediatr 1989;1 14:225-30. 142. Dame T, Gruters A, Schuppan D, Quantas N, Enders I, Weber B. Relationship of procollagen type 111 propeptide - related antigen in serum to somatic growth in healthy children and patients with growth disorders. J Pediatr 1989;1 14:257-60. 143. Carey DE, Goldberg B,Ratzan SK, Rubin KR, Rowe DW. Radioimmunoassay for type 1 procollagen in growth hormone-deficient children before and during treatment with growth hormone. Ped Res 1985;19:8-1 1. 144. Hyams, JS, Carey DE, Lelchtner AM, Goldberg BD. Type I procollagen as a biochemical marker of growth in children with inflammatory bowel disease. J Pediatr 1986; 109:619-24. 145. Tapanalnen P, Ristell L, Knip M, Kemppalnen L, K l l r M-L, Rlsteli J. Collagen metabolities in prediction of response to growth hormone (GH) therapy in short children. Horm Res 1990;33,Suppl 3:20. 146. Delmas PD, Malaval L, Arcot ME. Meunier PJ. Serum bone Gla-protein compared to bone histomorphometry in endocrine diseases. Bone 1985;6:339-41. 147. Johansen JS, Giwercman A, Hartwell D, et al. Serum bone Gla-protein as a marker of bone growth in children and adolescent$: correlation with age, height, serum insulin-like growth factor I and serum testosterone. J Clin Endocrinol1988;67:273-8. 148. Johansen JS, Jensen SB, R u s BJ, Rasmussen L, Zachrnann M, Christiansen C. Serum bone Gla-protein: a potential marker of growth hormone (GH) deficiency and the response to GH-therapy. J Clin Endocrinol Metab 1990;71:122-6.

Evaluation of growth hormone secretion and treatment.

The secretion of growth hormone (GH) is regulated by a complex system that includes both neurotransmitters and feedback by hormonal and metabolic subs...
1MB Sizes 0 Downloads 0 Views