Skip to main content
Erschienen in: Pediatric Nephrology 4/2019

Open Access 19.12.2017 | Review

Effects of long-term cysteamine treatment in patients with cystinosis

verfasst von: Gema Ariceta, Vincenzo Giordano, Fernando Santos

Erschienen in: Pediatric Nephrology | Ausgabe 4/2019

Abstract

Cystinosis is a rare autosomal-recessive lysosomal storage disease with high morbidity and mortality. It is caused by mutations in the CTNS gene that encodes the cystine transporter, cystinosin, which leads to lysosomal cystine accumulation. Patients with infantile nephropathic cystinosis, the most common and most severe clinical form of cystinosis, commonly present with renal Fanconi syndrome by 6–12 months of age, and without specific treatment, almost all will develop end-stage renal disease (ESRD) by 10–12 years of age. Early corneal cystine crystal deposition is a hallmark of the disease. Cystinosis also presents with gastrointestinal symptoms (e.g., vomiting, decreased appetite, and feeding difficulties) and severe growth retardation and may affect several other organs over time, including the eye, thyroid gland, gonads, pancreas, muscles, bone marrow, liver, nervous system, lungs, and bones. Cystine-depleting therapy with cysteamine orally is the only specific targeted therapy available for managing cystinosis and needs to be combined with cysteamine eye drops for corneal disease involvement. In patients with early treatment initiation and good compliance to therapy, long-term cysteamine treatment delays progression to ESRD, significantly improves growth, decreases the frequency and severity of extrarenal complications, and is associated with extended life expectancy. Therefore, early diagnosis of cystinosis and adequate life-long treatment with cysteamine are essential for preventing end-organ damage and improving the overall prognosis in these patients.

Introduction

Cystinosis is a rare autosomal-recessive lysosomal storage disease occurring approximately once every 100,000–200,000 live births, that is associated with high morbidity and mortality [1, 2]. The disease is caused by mutations in the CTNS gene that encodes the lysosomal cystine transporter, cystinosin, which result in the accumulation of cystine within the lysosome [3, 4]. There are three clinical forms of cystinosis: infantile or early-onset nephropathic cystinosis (OMIM 219800), juvenile or late-onset nephropathic cystinosis (OMIM 219900), and adult or ocular cystinosis (OMIM 219750) [1, 2]. Infantile nephropathic cystinosis is the most severe and most common form (~95% of cases) and that in which apparently healthy neonates usually present with renal Fanconi syndrome within the first year of life. In the absence of appropriate treatment, these patients develop end-stage renal disease (ESRD) by 10–12 years of age [5, 6]. The availability of renal replacement therapy (RRT) (i.e., dialysis and renal transplantation) since the early 1980s has improved the prognosis of patients with nephropathic cystinosis [7], but longer patient survival has highlighted the emergence of late manifestations of the disease. These extrarenal complications may develop in several organs, including the eye, thyroid gland, pancreas, gonads, lungs, muscles, brain, and bones [813]. Early diagnosis and adequate treatment is essential to prevent or attenuate end-organ damage and improve overall prognosis [14].
Life-long cystine-depleting therapy with cysteamine orally is the mainstay of treatment for cystinosis, and if initiated early, it can dramatically improve renal survival, patient’s quality of life, and overall disease prognosis [1, 15]. The most commonly used formulation is immediate-release cysteamine bitartrate (Cystagon®, Mylan Pharma, USA, and Orphan Europe, France), which has been on the market since the 1990s, and delayed-release cysteamine bitartrate (Procysbi®, Horizon Pharma, USA) was recently approved. Early and continuous long-term treatment with cysteamine is associated with delayed ESRD onset, a decreased risk of extrarenal complications, and improved survival rates [16, 17]. Frequency and severity of complications are decreased in patients with good compliance to cysteamine therapy [18, 19]. However, adolescents and adults tend to show lower treatment adherence after transition from pediatric to adult medical care. Given that the life expectancy of patients with cystinosis has now been increased into adulthood, as yet uncharacterized complications may emerge in well-treated adult patients with cystinosis [17].
Here we provide a brief overview of the pathogenesis of cystinosis and describe the long-term benefits of sustained cysteamine therapy, including reduced renal and extrarenal complications and increased life expectancy in patients with cystinosis.

Pathogenesis of cystinosis

Cystinosis is caused by bi-allelic mutations in the CTNS gene located on chromosome 17p13.2 that encodes for cystinosin, a ubiquitously expressed membrane protein that mediates the efflux of lysosomal cystine [3, 4, 20]. The most common CTNS mutation in Western populations is a large ~57-kb deletion that includes the D17S829 locus [3, 4], as well as the CARKL gene that encodes the enzyme sedoheptulokinase [21]. Nephropathic cystinosis is usually associated with two severe or truncating CTNS mutations that affect the promoter, leader sequence, transmembrane or non-transmembrane regions, including small deletions/insertions, missense and splicing mutations [1].
Although the exact mechanism of cystinosis is not fully understood, the lack of functional cystinosin is known to cause accumulation and crystallization of cystine within the lysosome, which ultimately results in apoptosis and tissue damage in all organ systems [22, 23]. Lysosomal cystine crystals are thought to cause elevated intracellular oxidative stress [24] and may activate inflammasome-related gene expression in proximal tubular epithelial cells [25]. In addition to cystine accumulation, studies have indicated that changes in the expression of transcription factors [26] and the mammalian target of rapamycin signaling pathway [27] may also play a role in the pathogenesis of cystinosis. Other mechanisms, such as the expression of abnormal cystinosin protein itself, increased apoptosis, proximal tubular cell metaplasia, and damage to podocytes, are also likely to play a role in the associated renal impairment[23, 28].

Cysteamine therapy

Cysteamine enters the lysosome by an unknown transporter and breaks down cystine into cysteine and cysteine–cysteamine disulfide, which are removed by the cysteine transporter and the lysine/arginine (PQLC2) transporter, respectively (Fig. 1) [29, 30]. Cysteamine rapidly deplete cells and tissues of lysosomal cystine [31] and is the only specific targeted therapy available for patients with cystinosis [1]. Based on the efficacy of orally administered cysteamine in slowing renal function decline [6, 32], the drug was approved for clinical use in the 1990s.
In vitro, cysteamine is associated with restoration of glutathione redox status in cultured cystinotic renal tubular epithelial cells [33] and differentiation of mesenchymal stromal bone marrow cells [34]. Cysteamine also increases spermatogenesis [35], reduces oxidative stress [36, 37], and prevents inhibition of thiol-containing enzymes [3840].
Cysteamine dosage calculations are based on body surface area (BSA) in adults, with a maximum dose of 1.95 g/m2 per day [17]. Intracellular leukocyte cystine (ILC) levels, which are typically >2 nmol half-cystine/mg protein in patients with cystinosis [1], may be monitored to optimize cysteamine dose and help identify patients with poor compliance. Target ILC trough levels of <1 nmol half-cystine/mg protein are used for adequate disease control and cysteamine dose adjustment [6]. However, it should be noted that ILC levels can vary between laboratories, and the exact benefits of measuring levels is not well established.
At higher doses (>1.95 g/m2 per day) of cysteamine, rare adverse effects have been reported, including bone pain, myalgia, skin striae, and bruise-like lesions on the elbows (reactive angioendotheliomatosis on skin biopsy) [41]. Cysteamine increases human dermal microvascular endothelial cell survival in vitro, which may explain how the drug causes angioendotheliomatosis [42]. As it is metabolized into volatile sulfur compounds (i.e., dimethyl sulfide and methanethiol), treatment is also associated with an unpleasant sulfurous body and breath odor, which can often lead to poor treatment compliance [41].
Other cysteamine-related adverse events include allergic rash, hyperthermia, lethargy, neutropenia, seizures, and gastrointestinal discomfort; however, these effects are reversible, and most are prevented by starting the drug at a low dosage with gradual titration as tolerated under the guidance of experienced physicians [43]. As cysteamine toxicity may be associated with copper deficiency in patients with renal Fanconi syndrome, copper supplementation may prevent cysteamine-related adverse effects in some patients [44].

Long-term effects of cysteamine therapy

Long-term cysteamine treatment orally is associated with improvements in renal and extrarenal complications. A summary of clinical data showing the long-term effects ofshown in Table 1. Where reported [5, 45, 47], the duration of follow-up ranged from 8 to 25 years, and the largest study comprised data from 208 patients [49]. Of note, follow-up with early treatment initiation is currently limited to <30 years; therefore, the effects of cysteamine in older patients are unknown. It should also be noted that, in some studies, the lack of adequate cysteamine therapy may be due to insufficient access to treatment (e.g., in developing countries), as well as poor treatment compliance.
Table 1
Clinical data showing the long-term effects of cysteamine therapy orally in patients with cystinosis
First author, year
Study description
Renal outcomes
Extrarenal complications
Brodin-Sartorius, 2012 [45]
French study; adults (aged ≥15 years) diagnosed between 1961 and 1995; mean follow-up 24.6 years (n = 86)
• Initiation <5 years of age vs later delayed ESRD onset (mean age at onset 13.4 vs 9.6 years; p < 0.05)
• Increased treatment compliance also delayed ESRD onset (p < 0.0001)
• Initiation <5 years of age (vs later or no treatment) reduced rates of hypothyroidism (52.5% vs 73.3% and 96.8%), diabetes (27.5% vs 64.7% and 89.7%), neuromuscular disorders (15% vs 53.6% and 61.1%), and death (5% vs 42.9% and 63.6%) [p < 0.001 for each parameter]
Broyer, 2008 [46]
Necker Enfants-Malades Hospital series; patients born before 1988; aged 20–39 years (n = 56)
• Initiation <3 years of age vs later delayed ESRD onset (mean age at onset 17.4 vs 9.6 years)
• Initiation <3 years of age vs later:
- Improved linear growth (mean height 167 vs 144.5 cm for boys; 153.5 vs 132 cm for girls) and visual acuity (mean 8.5 vs 5)
• Reduced rates of glucose intolerance, thyroxine requirements (35.7% vs 88.2%), myopathy (0% vs 38.5%), cerebellar/pyramidal symptoms or mental deterioration (0% vs 35.9%), and hepatosplenic disorders (0% vs 92.3%)
Gahl, 2007 [16]
US database study; adults (aged 18–45 years) examined between January 1985 and May 2006 (n = 100)
• Treatment for ≥8 years vs <8 years delayed renal transplantation (mean age 14.8 vs 11.0 years)
• Increased duration of therapy (from <10 to >20 years) reduced rates of diabetes (from 28 to 0%), myopathy (from 60 to 0%), pulmonary dysfunction (from 80 to 0%), and death (from 43 to 0%)
• Treatment for ≥8 vs <8 years significantly improved mean height (154.7 vs 143.6 cm) and weight (53.2 vs 45.3 kg) parameters, and rates of hypothyroidism (56% vs 87%) and death (8% vs 49%)
Greco, 2010 [5]
Italian single-center study; patients diagnosed at 3–60 years of age; median follow-up 17.6 years (n = 23)
• Initiation <2.5 years of age vs later improved evolution of renal function (p = 0.006), and decreased risk of CKD stage 3
• Rapid renal function decline prevented by higher cysteamine doses and use of ACEi
• Patients treated more recently (initiated <2.5 years of age) had improved linear growth curves vs older children (p = 0.04)
Gultekingil Keser, 2014 [47]
Turkish, retrospective, single-center study; patients aged 0.5–29 years; median follow-up 8 years (n = 21)
NR
• Patients with ILC levels <2 vs ≥2 nmol half-cystine/mg protein had reduced rates of short stature (66.6% vs 90.9%), pubertal delay (0% vs 66.6%), hypothyroidism (33.3% vs 54.5%), and diabetes (0% vs 18.1%)
Nesterova, 2015 [19]
US database study; patients aged 11–48 years examined between 1975 and 2005 (n = 147)
• Treatment compliance was linearly correlated with age at ESRD onset (R2 = 0.997)
• Optimal treatment for 1 year preserved 0.9 years of renal glomerular function
• Severity of renal Fanconi syndrome (decreased tubular function) was not affected
NR
Sonies, 2005 [48]
US database study; patients aged 6–45 years examined between February 1987 and March 2004 (n = 101)
NR
• Severity of swallowing dysfunction (muscle atrophy) decreased with increased number of years of treatment
Tsilou, 2006 [49]
US database study; patients aged 0.5–42 years examined between 1976 and 2004 (n = 208)
NR
• Treatment for >20 vs ≤10 years decreased rate of retinopathy (0% vs 28%)
Vaisbich, 2010 [50]
Brazilian Multicenter Nephropathic Study; patients aged 1.3–29 years enrolled since 1999 (n = 102)
• Initiation <2 years of age vs later reduced rate of CKD stage II–V (25% vs 77.5%)
• Initiation <2 years vs later:
• Improved growth (weight and height) parameters
• Reduced rates of hypothyroidism (10% vs 61.7%), diabetes (5% vs 7.8%), muscular weakness (0% vs 6.8%), hepatic dysfunction (0% vs 4.9%), CNS disorders (0% vs 4.9%), and swallowing dysfunction (0% vs 1.9%)
Viltz, 2013 [51]
Children and adolescents (aged 3–18 years) [n = 46]
NR
• Initiation <2 years vs later improved cognitive function (verbal, performance, and full-scale IQ scores, and spatial-relations test; p < 0.05), but not visual–motor performance scores
ACEi angiotensin-converting enzyme inhibitor, CKD chronic kidney disease, CNS central nervous system, ESRD end-stage renal disease, ILC intracellular leukocyte cystine, IQ intelligence quotient, NR not reported

Renal outcomes

Although cysteamine does not significantly affect renal Fanconi syndrome in most patients [1, 2], in patients with early initiation and good treatment compliance, long-term cysteamine therapy improves the evolution of renal function in patients with cystinosis, delaying progression of glomerular function deterioration, ESRD onset, and the need for renal transplantation [5, 16, 19, 45, 46, 50]. Early initiation was associated with preserved renal function and reduced incidence of chronic kidney disease in both a Brazilian multicenter study [50] and a long-term retrospective study [5]. In the latter study, higher doses of cysteamine and the use of angiotensin-converting enzyme inhibitors (ACEis) were also significantly associated with a delay in the decline of renal function [5]. Early initiation and adequate long-term cysteamine therapy delayed the onset of ESRD in the Necker-Enfants Malades Hospital series of patients with cystinosis, who survived to at least 20 years of age [46], and in a French study of adults [45]. Increased compliance to cysteamine therapy also had a linear correlation with the mean age at ESRD onset (R2 = 0.997) in a long-term retrospective study [19].
Taken together, these studies confirm that cysteamine therapy orally must be initiated as early as possible following diagnosis, even in small infants, and be used long term with good compliance to maximize its beneficial effects on delaying progression of renal failure and ESRD onset.

Extrarenal complications

In long-term studies, patients who initiated treatment early [5, 45, 46, 50, 51] and received adequate cysteamine therapy [16] had increased life expectancy and fewer complications than inadequately treated patients and those who started treatment later or were untreated (Table 1). Given the strong evidence for cysteamine-associated benefits and longer life expectancy in most patients, there is increasing interest in improving patient care with regard to extrarenal disease manifestations, treatment adherence, and adult patient care.

Eye disorders

Untreated patients with infantile nephropathic cystinosis develop corneal cystine crystal accumulation that leads to ocular discomfort and photophobia [1, 13]. As cysteamine therapy orally has no effect on corneal cystine crystals, cysteamine eye drops are used to dissolve crystals and reduce symptoms. A new topical viscous formulation of cysteamine eye drops has been developed that requires less frequent instillation and is more effective than standard cysteamine eye drops [52].
Intracellular cystine crystal deposition can also occur in posterior-segment ocular structures and is associated with damage to retina, choroid, and optic nerve [13, 49]. Long-term cysteamine therapy orally may reduce posterior-segment complications. In a retrospective US National Institutes of Health (NIH) study, a decreased incidence of retinopathy from 28 to 0% was observed among patients with cystinosis when therapy was increased from ≤10 years to 21–30 years [49].

Endocrine disorders

Long-term cysteamine therapy orally is associated with marked improvements in growth retardation and other endocrine disorders [5, 16, 46, 47, 50]. Growth hormone therapy may also improve growth outcomes [1]. Early initiation and long-term therapy led to significantly improved mean height [16, 46] and weight [16, 50] parameters in several studies. Early initiation was also associated with improved linear growth curves in an Italian study, where poorer linear growth was observed in older children (in whom cysteamine was started after 2.5 years of age and suboptimal treatment was more likely) compared with those who initiated cysteamine more recently [5]. Early therapy initiation is also associated with a lower incidence of hypothyroidism and diabetes mellitus [46, 50] and significant delays in the onset of these conditions [45]. Furthermore, long-term (≥8 years) cysteamine therapy reduces the incidence of hypothyroidism and diabetes in adults with cystinosis [16].
A long-term study (median follow-up 8 years) indicated that good compliance to cysteamine therapy orally is effective in preventing endocrine disorders in patients with cystinosis, with ILC levels of <2 nmol half-cystine/mg protein being associated with lower incidences of short stature, pubertal delay, hypothyroidism, and diabetes than those with higher ILC levels [47].

Neurocognitive disorders

Cystinosis is associated with several neurologic complications, including structural brain abnormalities, cognitive impairment, and learning difficulties [8]. Long-term studies have indicated that early initiation of cysteamine orally may prevent many of these neurocognitive disorders [46, 50, 51]. Cerebellar or pyramidal symptoms or mental deterioration [46] or central nervous system disorders [50] were only observed among patients who started cysteamine after 2 [50] or 3 [46] years of age. Early initiation (<2 years of age) is also associated with significantly higher performance, verbal, and full-scale IQ scores, and significantly improved visual–spatial skills, although visual–motor skills were not significantly affected [51].

Muscular disorders

Distal myopathy and muscle atrophy are significant complications of cystinosis among adolescents and young adults, particularly in untreated patients and those with poor compliance to cysteamine therapy [9]. The frequency and severity of these complications can be reduced with long-term cysteamine, if initiated early. In adults, long-term (>20 years) therapy was associated with a decrease in the incidence of myopathy (to 0%) compared with a treatment duration of ≤10 years (60%) [16], and the incidence of neuromuscular complications decreased significantly among patients who initiated therapy early (<5 years) [45]. In two studies, oropharyngeal dysfunction and/or distal muscle atrophy were only found in patients who started therapy >2 [50] or 3 [46] years of age. Swallowing dysfunction, which occurs due to muscle atrophy and increases the risk of fatal aspiration, decreased in severity with increased duration of treatment [48].

Pulmonary disorders

Patients with cystinosis may develop pulmonary complications, including respiratory failure caused by muscle weakness, sleep apnea, and aspiration due to swallowing dysfunction [10]. In adults, longer-term cysteamine therapy is associated with a lower incidence of pulmonary disorders [16]. Mean forced vital capacity was higher among patients who received cysteamine for >10 years (83% of predicted) than in those with ≤10 years of therapy (54% of predicted) [16].

Mortality

Long-term cysteamine therapy is associated with improved survival rates among patients with cystinosis, particularly when initiated early [16, 45, 46]. In adults, long-term cysteamine was associated with lower mortality rates, with the incidence of death decreasing as the duration of therapy increased [16]; in other studies, lower rates of mortality were observed among patients who started therapy early (<3 [46] or 5 [45] years).

Treatment adherence

The benefits of long-term cysteamine therapy are most evident in patients with good compliance to treatment and are related to the duration of therapy [18, 19]. In a retrospective NIH study, each year of excellent cystine-depleting therapy resulted in almost 1 year of preserved renal function [19]. Treatment tends to be most successful in children aged <11 years, as they have the highest treatment adherence, largely due to administration of cysteamine being conducted by a parent [18].

Conclusions

Inadequately treated cystinosis can lead to multisystem morbidity and mortality. However, long-term cysteamine therapy, if initiated early and used with good treatment compliance, provides significant improvements in renal and extrarenal outcomes. Early diagnosis and adequate long-term treatment are therefore essential for preventing end-organ damage and improving the overall prognosis in these patients. Major efforts to maintain and promote adequate long-term cysteamine therapy should be undertaken by healthcare professionals, patients, and supportive groups.

Acknowledgements

The authors would like to thank Sarah Greig, PhD, of Springer Healthcare Communications, who prepared the outline and subsequent drafts of this manuscript, and assisted with revisions after peer review. This medical writing assistance was funded by Orphan Europe.

Compliance with ethical standards

Conflicts of interest/financial disclosures

The authors declare no conflict of interest. VG is an Orphan Europe employee.
Open Access This article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://creativecommons.org/licenses/by/4.0/), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made.

Unsere Produktempfehlungen

e.Med Interdisziplinär

Kombi-Abonnement

Für Ihren Erfolg in Klinik und Praxis - Die beste Hilfe in Ihrem Arbeitsalltag

Mit e.Med Interdisziplinär erhalten Sie Zugang zu allen CME-Fortbildungen und Fachzeitschriften auf SpringerMedizin.de.

e.Med Pädiatrie

Kombi-Abonnement

Mit e.Med Pädiatrie erhalten Sie Zugang zu CME-Fortbildungen des Fachgebietes Pädiatrie, den Premium-Inhalten der pädiatrischen Fachzeitschriften, inklusive einer gedruckten Pädiatrie-Zeitschrift Ihrer Wahl.

e.Med Urologie

Kombi-Abonnement

Mit e.Med Urologie erhalten Sie Zugang zu den urologischen CME-Fortbildungen und Premium-Inhalten der urologischen Fachzeitschriften.

Literatur
2.
Zurück zum Zitat Emma F, Nesterova G, Langman C, Labbe A, Cherqui S, Goodyer P, Janssen MC, Greco M, Topaloglu R, Elenberg E, Dohil R, Trauner D, Antignac C, Cochat P, Kaskel F, Servais A, Wuhl E, Niaudet P, Van't Hoff W, Gahl W, Levtchenko E (2014) Nephropathic cystinosis: an international consensus document. Nephrol Dial Transplant 29(Suppl 4):iv87-94CrossRefPubMed Emma F, Nesterova G, Langman C, Labbe A, Cherqui S, Goodyer P, Janssen MC, Greco M, Topaloglu R, Elenberg E, Dohil R, Trauner D, Antignac C, Cochat P, Kaskel F, Servais A, Wuhl E, Niaudet P, Van't Hoff W, Gahl W, Levtchenko E (2014) Nephropathic cystinosis: an international consensus document. Nephrol Dial Transplant 29(Suppl 4):iv87-94CrossRefPubMed
3.
Zurück zum Zitat Thoene J, Lemons R, Anikster Y, Mullet J, Paelicke K, Lucero C, Gahl W, Schneider J, Shu SG, Campbell HT (1999) Mutations of CTNS causing intermediate cystinosis. Mol Genet Metab 67:283–293CrossRefPubMed Thoene J, Lemons R, Anikster Y, Mullet J, Paelicke K, Lucero C, Gahl W, Schneider J, Shu SG, Campbell HT (1999) Mutations of CTNS causing intermediate cystinosis. Mol Genet Metab 67:283–293CrossRefPubMed
4.
Zurück zum Zitat Town M, Jean G, Cherqui S, Attard M, Forestier L, Whitmore SA, Callen DF, Gribouval O, Broyer M, Bates GP, van’t Hoff W, Antignac C (1998) A novel gene encoding an integral membrane protein is mutated in nephropathic cystinosis. Nat Genet 18:319–324CrossRefPubMed Town M, Jean G, Cherqui S, Attard M, Forestier L, Whitmore SA, Callen DF, Gribouval O, Broyer M, Bates GP, van’t Hoff W, Antignac C (1998) A novel gene encoding an integral membrane protein is mutated in nephropathic cystinosis. Nat Genet 18:319–324CrossRefPubMed
5.
Zurück zum Zitat Greco M, Brugnara M, Zaffanello M, Taranta A, Pastore A, Emma F (2010) Long-term outcome of nephropathic cystinosis: a 20-year single-center experience. Pediatr Nephrol 25:2459–2467CrossRefPubMed Greco M, Brugnara M, Zaffanello M, Taranta A, Pastore A, Emma F (2010) Long-term outcome of nephropathic cystinosis: a 20-year single-center experience. Pediatr Nephrol 25:2459–2467CrossRefPubMed
6.
Zurück zum Zitat Markello TC, Bernardini IM, Gahl WA (1993) Improved renal function in children with cystinosis treated with cysteamine. N Engl J Med 328:1157–1162CrossRefPubMed Markello TC, Bernardini IM, Gahl WA (1993) Improved renal function in children with cystinosis treated with cysteamine. N Engl J Med 328:1157–1162CrossRefPubMed
7.
Zurück zum Zitat Van Stralen KJ, Emma F, Jager KJ, Verrina E, Schaefer F, Laube GF, Lewis MA, Levtchenko EN (2011) Improvement in the renal prognosis in nephropathic cystinosis. Clin J Am Soc Nephrol 6:2485–2491CrossRefPubMedPubMedCentral Van Stralen KJ, Emma F, Jager KJ, Verrina E, Schaefer F, Laube GF, Lewis MA, Levtchenko EN (2011) Improvement in the renal prognosis in nephropathic cystinosis. Clin J Am Soc Nephrol 6:2485–2491CrossRefPubMedPubMedCentral
8.
9.
Zurück zum Zitat Thoene JG (2017) Myopathy and less frequent complications of cystinosis. J Pediatr 183S:S22–S23CrossRefPubMed Thoene JG (2017) Myopathy and less frequent complications of cystinosis. J Pediatr 183S:S22–S23CrossRefPubMed
11.
13.
Zurück zum Zitat Bishop R (2017) Ocular complications of infantile nephropathic cystinosis. J Pediatr 183S:S19–S21CrossRefPubMed Bishop R (2017) Ocular complications of infantile nephropathic cystinosis. J Pediatr 183S:S19–S21CrossRefPubMed
14.
Zurück zum Zitat Gahl WA (2003) Early oral cysteamine therapy for nephropathic cystinosis. Eur J Pediatr 162(Suppl 1):S38–S41CrossRefPubMed Gahl WA (2003) Early oral cysteamine therapy for nephropathic cystinosis. Eur J Pediatr 162(Suppl 1):S38–S41CrossRefPubMed
15.
Zurück zum Zitat Kleta R, Bernardini I, Ueda M, Varade WS, Phornphutkul C, Krasnewich D, Gahl WA (2004) Long-term follow-up of well-treated nephropathic cystinosis patients. J Pediatr 145:555–560CrossRefPubMed Kleta R, Bernardini I, Ueda M, Varade WS, Phornphutkul C, Krasnewich D, Gahl WA (2004) Long-term follow-up of well-treated nephropathic cystinosis patients. J Pediatr 145:555–560CrossRefPubMed
16.
Zurück zum Zitat Gahl WA, Balog JZ, Kleta R (2007) Nephropathic cystinosis in adults: natural history and effects of oral cysteamine therapy. Ann Intern Med 147:242–250CrossRefPubMed Gahl WA, Balog JZ, Kleta R (2007) Nephropathic cystinosis in adults: natural history and effects of oral cysteamine therapy. Ann Intern Med 147:242–250CrossRefPubMed
17.
Zurück zum Zitat Langman CB, Barshop BA, Deschenes G, Emma F, Goodyer P, Lipkin G, Midgley JP, Ottolenghi C, Servais A, Soliman NA, Thoene JG, Levtchenko EN (2016) Controversies and research agenda in nephropathic cystinosis: conclusions from a “kidney disease: improving global outcomes” (KDIGO) controversies conference. Kidney Int 89:1192–1203CrossRefPubMed Langman CB, Barshop BA, Deschenes G, Emma F, Goodyer P, Lipkin G, Midgley JP, Ottolenghi C, Servais A, Soliman NA, Thoene JG, Levtchenko EN (2016) Controversies and research agenda in nephropathic cystinosis: conclusions from a “kidney disease: improving global outcomes” (KDIGO) controversies conference. Kidney Int 89:1192–1203CrossRefPubMed
18.
Zurück zum Zitat Ariceta G, Lara E, Camacho JA, Oppenheimer F, Vara J, Santos F, Munoz MA, Cantarell C, Gil Calvo M, Romero R, Valenciano B, Garcia-Nieto V, Sanahuja MJ, Crespo J, Justa ML, Urisarri A, Bedoya R, Bueno A, Daza A, Bravo J, Llamas F, Jimenez Del Cerro LA (2015) Cysteamine (Cystagon®) adherence in patients with cystinosis in Spain: successful in children and a challenge in adolescents and adults. Nephrol Dial Transplant 30:475–480CrossRefPubMed Ariceta G, Lara E, Camacho JA, Oppenheimer F, Vara J, Santos F, Munoz MA, Cantarell C, Gil Calvo M, Romero R, Valenciano B, Garcia-Nieto V, Sanahuja MJ, Crespo J, Justa ML, Urisarri A, Bedoya R, Bueno A, Daza A, Bravo J, Llamas F, Jimenez Del Cerro LA (2015) Cysteamine (Cystagon®) adherence in patients with cystinosis in Spain: successful in children and a challenge in adolescents and adults. Nephrol Dial Transplant 30:475–480CrossRefPubMed
19.
Zurück zum Zitat Nesterova G, Williams C, Bernardini I, Gahl WA (2015) Cystinosis: renal glomerular and renal tubular function in relation to compliance with cystine-depleting therapy. Pediatr Nephrol 30:945–951CrossRefPubMed Nesterova G, Williams C, Bernardini I, Gahl WA (2015) Cystinosis: renal glomerular and renal tubular function in relation to compliance with cystine-depleting therapy. Pediatr Nephrol 30:945–951CrossRefPubMed
20.
Zurück zum Zitat McDowell GA, Gahl WA, Stephenson LA, Schneider JA, Weissenbach J, Polymeropoulos MH, Town MM, van't Hoff W, Farrall M, Mathew CG (1995) Linkage of the gene for cystinosis to markers on the short arm of chromosome 17. Nat Genet 10:246–248CrossRef McDowell GA, Gahl WA, Stephenson LA, Schneider JA, Weissenbach J, Polymeropoulos MH, Town MM, van't Hoff W, Farrall M, Mathew CG (1995) Linkage of the gene for cystinosis to markers on the short arm of chromosome 17. Nat Genet 10:246–248CrossRef
21.
Zurück zum Zitat Wamelink MM, Struys EA, Jansen EE, Levtchenko EN, Zijlstra FS, Engelke U, Blom HJ, Jakobs C, Wevers RA (2008) Sedoheptulokinase deficiency due to a 57-kb deletion in cystinosis patients causes urinary accumulation of sedoheptulose: elucidation of the CARKL gene. Hum Mutat 29:532–536CrossRefPubMed Wamelink MM, Struys EA, Jansen EE, Levtchenko EN, Zijlstra FS, Engelke U, Blom HJ, Jakobs C, Wevers RA (2008) Sedoheptulokinase deficiency due to a 57-kb deletion in cystinosis patients causes urinary accumulation of sedoheptulose: elucidation of the CARKL gene. Hum Mutat 29:532–536CrossRefPubMed
22.
Zurück zum Zitat Park M, Helip-Wooley A, Thoene J (2002) Lysosomal cystine storage augments apoptosis in cultured human fibroblasts and renal tubular epithelial cells. J Am Soc Nephrol 13:2878–2887CrossRefPubMed Park M, Helip-Wooley A, Thoene J (2002) Lysosomal cystine storage augments apoptosis in cultured human fibroblasts and renal tubular epithelial cells. J Am Soc Nephrol 13:2878–2887CrossRefPubMed
23.
Zurück zum Zitat Cherqui S, Courtoy PJ (2017) The renal Fanconi syndrome in cystinosis: pathogenic insights and therapeutic perspectives. Nat Rev Nephrol 13:115–131CrossRefPubMed Cherqui S, Courtoy PJ (2017) The renal Fanconi syndrome in cystinosis: pathogenic insights and therapeutic perspectives. Nat Rev Nephrol 13:115–131CrossRefPubMed
24.
Zurück zum Zitat Sumayao R, McEvoy B, Newsholme P, McMorrow T (2016) Lysosomal cystine accumulation promotes mitochondrial depolarization and induction of redox-sensitive genes in human kidney proximal tubular cells. J Physiol 594:3353–3370CrossRefPubMedPubMedCentral Sumayao R, McEvoy B, Newsholme P, McMorrow T (2016) Lysosomal cystine accumulation promotes mitochondrial depolarization and induction of redox-sensitive genes in human kidney proximal tubular cells. J Physiol 594:3353–3370CrossRefPubMedPubMedCentral
25.
Zurück zum Zitat Prencipe G, Caiello I, Cherqui S, Whisenant T, Petrini S, Emma F, De Benedetti F (2014) Inflammasome activation by cystine crystals: implications for the pathogenesis of cystinosis. J Am Soc Nephrol 25:1163–1169CrossRefPubMedPubMedCentral Prencipe G, Caiello I, Cherqui S, Whisenant T, Petrini S, Emma F, De Benedetti F (2014) Inflammasome activation by cystine crystals: implications for the pathogenesis of cystinosis. J Am Soc Nephrol 25:1163–1169CrossRefPubMedPubMedCentral
26.
Zurück zum Zitat Rega LR, Polishchuk E, Montefusco S, Napolitano G, Tozzi G, Zhang J, Bellomo F, Taranta A, Pastore A, Polishchuk R, Piemonte F, Medina DL, Catz SD, Ballabio A, Emma F (2016) Activation of the transcription factor EB rescues lysosomal abnormalities in cystinotic kidney cells. Kidney Int 89:862–873CrossRefPubMed Rega LR, Polishchuk E, Montefusco S, Napolitano G, Tozzi G, Zhang J, Bellomo F, Taranta A, Pastore A, Polishchuk R, Piemonte F, Medina DL, Catz SD, Ballabio A, Emma F (2016) Activation of the transcription factor EB rescues lysosomal abnormalities in cystinotic kidney cells. Kidney Int 89:862–873CrossRefPubMed
27.
Zurück zum Zitat Andrzejewska Z, Nevo N, Thomas L, Chhuon C, Bailleux A, Chauvet V, Courtoy PJ, Chol M, Guerrera IC, Antignac C (2016) Cystinosin is a component of the vacuolar H+−ATPase-Ragulator-rag complex controlling mammalian target of rapamycin complex 1 signaling. J Am Soc Nephrol 27:1678–1688CrossRefPubMed Andrzejewska Z, Nevo N, Thomas L, Chhuon C, Bailleux A, Chauvet V, Courtoy PJ, Chol M, Guerrera IC, Antignac C (2016) Cystinosin is a component of the vacuolar H+−ATPase-Ragulator-rag complex controlling mammalian target of rapamycin complex 1 signaling. J Am Soc Nephrol 27:1678–1688CrossRefPubMed
28.
Zurück zum Zitat Ivanova EA, Arcolino FO, Elmonem MA, Rastaldi MP, Giardino L, Cornelissen EM, van den Heuvel LP, Levtchenko EN (2016) Cystinosin deficiency causes podocyte damage and loss associated with increased cell motility. Kidney Int 89:1037–1048CrossRefPubMed Ivanova EA, Arcolino FO, Elmonem MA, Rastaldi MP, Giardino L, Cornelissen EM, van den Heuvel LP, Levtchenko EN (2016) Cystinosin deficiency causes podocyte damage and loss associated with increased cell motility. Kidney Int 89:1037–1048CrossRefPubMed
29.
Zurück zum Zitat Jezegou A, Llinares E, Anne C, Kieffer-Jaquinod S, O'Regan S, Aupetit J, Chabli A, Sagne C, Debacker C, Chadefaux-Vekemans B, Journet A, Andre B, Gasnier B (2012) Heptahelical protein PQLC2 is a lysosomal cationic amino acid exporter underlying the action of cysteamine in cystinosis therapy. Proc Natl Acad Sci U S A 109:E3434–E3443CrossRefPubMedPubMedCentral Jezegou A, Llinares E, Anne C, Kieffer-Jaquinod S, O'Regan S, Aupetit J, Chabli A, Sagne C, Debacker C, Chadefaux-Vekemans B, Journet A, Andre B, Gasnier B (2012) Heptahelical protein PQLC2 is a lysosomal cationic amino acid exporter underlying the action of cysteamine in cystinosis therapy. Proc Natl Acad Sci U S A 109:E3434–E3443CrossRefPubMedPubMedCentral
30.
Zurück zum Zitat Pisoni RL, Thoene JG, Christensen HN (1985) Detection and characterization of carrier-mediated cationic amino acid transport in lysosomes of normal and cystinotic human fibroblasts. Role in therapeutic cystine removal? J Biol Chem 260:4791–4798PubMed Pisoni RL, Thoene JG, Christensen HN (1985) Detection and characterization of carrier-mediated cationic amino acid transport in lysosomes of normal and cystinotic human fibroblasts. Role in therapeutic cystine removal? J Biol Chem 260:4791–4798PubMed
31.
Zurück zum Zitat Thoene JG, Oshima RG, Crawhall JC, Olson DL, Schneider JA (1976) Cystinosis. Intracellular cystine depletion by aminothiols in vitro and in vivo. J Clin Invest 58:180–189CrossRefPubMedPubMedCentral Thoene JG, Oshima RG, Crawhall JC, Olson DL, Schneider JA (1976) Cystinosis. Intracellular cystine depletion by aminothiols in vitro and in vivo. J Clin Invest 58:180–189CrossRefPubMedPubMedCentral
32.
Zurück zum Zitat Gahl WA, Reed GF, Thoene JG, Schulman JD, Rizzo WB, Jonas AJ, Denman DW, Schlesselman JJ, Corden BJ, Schneider JA (1987) Cysteamine therapy for children with nephropathic cystinosis. N Engl J Med 316:971–977CrossRefPubMed Gahl WA, Reed GF, Thoene JG, Schulman JD, Rizzo WB, Jonas AJ, Denman DW, Schlesselman JJ, Corden BJ, Schneider JA (1987) Cysteamine therapy for children with nephropathic cystinosis. N Engl J Med 316:971–977CrossRefPubMed
33.
Zurück zum Zitat Wilmer MJ, Kluijtmans LA, van der Velden TJ, Willems PH, Scheffer PG, Masereeuw R, Monnens LA, van den Heuvel LP, Levtchenko EN (2011) Cysteamine restores glutathione redox status in cultured cystinotic proximal tubular epithelial cells. Biochim Biophys Acta 1812:643–651CrossRefPubMed Wilmer MJ, Kluijtmans LA, van der Velden TJ, Willems PH, Scheffer PG, Masereeuw R, Monnens LA, van den Heuvel LP, Levtchenko EN (2011) Cysteamine restores glutathione redox status in cultured cystinotic proximal tubular epithelial cells. Biochim Biophys Acta 1812:643–651CrossRefPubMed
34.
Zurück zum Zitat Conforti A, Taranta A, Biagini S, Starc N, Pitisci A, Bellomo F, Cirillo V, Locatelli F, Bernardo ME, Emma F (2015) Cysteamine treatment restores the in vitro ability to differentiate along the osteoblastic lineage of mesenchymal stromal cells isolated from bone marrow of a cystinotic patient. J Transl Med 13:143CrossRefPubMedPubMedCentral Conforti A, Taranta A, Biagini S, Starc N, Pitisci A, Bellomo F, Cirillo V, Locatelli F, Bernardo ME, Emma F (2015) Cysteamine treatment restores the in vitro ability to differentiate along the osteoblastic lineage of mesenchymal stromal cells isolated from bone marrow of a cystinotic patient. J Transl Med 13:143CrossRefPubMedPubMedCentral
36.
Zurück zum Zitat Okamura DM, Bahrami NM, Ren S, Pasichnyk K, Williams JM, Gangoiti JA, Lopez-Guisa JM, Yamaguchi I, Barshop BA, Duffield JS, Eddy AA (2014) Cysteamine modulates oxidative stress and blocks myofibroblast activity in CKD. J Am Soc Nephrol 25:43–54CrossRefPubMed Okamura DM, Bahrami NM, Ren S, Pasichnyk K, Williams JM, Gangoiti JA, Lopez-Guisa JM, Yamaguchi I, Barshop BA, Duffield JS, Eddy AA (2014) Cysteamine modulates oxidative stress and blocks myofibroblast activity in CKD. J Am Soc Nephrol 25:43–54CrossRefPubMed
37.
Zurück zum Zitat Kessler A, Biasibetti M, da Silva Melo DA, Wajner M, Dutra-Filho CS, de Souza Wyse AT, Wannmacher CM (2008) Antioxidant effect of cysteamine in brain cortex of young rats. Neurochem Res 33:737–744CrossRefPubMed Kessler A, Biasibetti M, da Silva Melo DA, Wajner M, Dutra-Filho CS, de Souza Wyse AT, Wannmacher CM (2008) Antioxidant effect of cysteamine in brain cortex of young rats. Neurochem Res 33:737–744CrossRefPubMed
38.
Zurück zum Zitat Fleck RM, Rodrigues Junior V, Giacomazzi J, Parissoto D, Dutra-Filho CS, de Souza Wyse AT, Wajner M, Wannmacher CM (2005) Cysteamine prevents and reverses the inhibition of creatine kinase activity caused by cystine in rat brain cortex. Neurochem Int 46:391–397CrossRefPubMed Fleck RM, Rodrigues Junior V, Giacomazzi J, Parissoto D, Dutra-Filho CS, de Souza Wyse AT, Wajner M, Wannmacher CM (2005) Cysteamine prevents and reverses the inhibition of creatine kinase activity caused by cystine in rat brain cortex. Neurochem Int 46:391–397CrossRefPubMed
39.
Zurück zum Zitat Rosa TG, de Souza Wyse AT, Wajner M, Wannmacher CM (2004) Cysteamine prevents and reverses the inhibition of pyruvate kinase activity caused by cystine in rat heart. Biochim Biophys Acta 1689:114–119CrossRefPubMed Rosa TG, de Souza Wyse AT, Wajner M, Wannmacher CM (2004) Cysteamine prevents and reverses the inhibition of pyruvate kinase activity caused by cystine in rat heart. Biochim Biophys Acta 1689:114–119CrossRefPubMed
40.
Zurück zum Zitat Rech VC, Feksa LR, Fleck RM, Athaydes GA, Dornelles PK, Rodrigues-Junior V, Wannmacher CM (2008) Cysteamine prevents inhibition of thiol-containing enzymes caused by cystine or cystine dimethylester loading in rat brain cortex. Metab Brain Dis 23:133–145CrossRefPubMed Rech VC, Feksa LR, Fleck RM, Athaydes GA, Dornelles PK, Rodrigues-Junior V, Wannmacher CM (2008) Cysteamine prevents inhibition of thiol-containing enzymes caused by cystine or cystine dimethylester loading in rat brain cortex. Metab Brain Dis 23:133–145CrossRefPubMed
41.
Zurück zum Zitat Besouw MT, Bowker R, Dutertre JP, Emma F, Gahl WA, Greco M, Lilien MR, McKiernan J, Nobili F, Schneider JA, Skovby F, van den Heuvel LP, Van't Hoff WG, Levtchenko EN (2011) Cysteamine toxicity in patients with cystinosis. J Pediatr 159:1004–1011CrossRefPubMed Besouw MT, Bowker R, Dutertre JP, Emma F, Gahl WA, Greco M, Lilien MR, McKiernan J, Nobili F, Schneider JA, Skovby F, van den Heuvel LP, Van't Hoff WG, Levtchenko EN (2011) Cysteamine toxicity in patients with cystinosis. J Pediatr 159:1004–1011CrossRefPubMed
42.
Zurück zum Zitat Besouw M, van den Heuvel L, van Eijsden R, Bongaers I, Kluijtmans L, Dewerchin M, Levtchenko E (2013) Increased human dermal microvascular endothelial cell survival induced by cysteamine. J Inherit Metab Dis 36:1073–1077CrossRefPubMed Besouw M, van den Heuvel L, van Eijsden R, Bongaers I, Kluijtmans L, Dewerchin M, Levtchenko E (2013) Increased human dermal microvascular endothelial cell survival induced by cysteamine. J Inherit Metab Dis 36:1073–1077CrossRefPubMed
43.
Zurück zum Zitat Corden BJ, Schulman JD, Schneider JA, Thoene JG (1981) Adverse reactions to oral cysteamine use in nephropathic cystinosis. Dev Pharmacol Ther 3:25–30CrossRefPubMed Corden BJ, Schulman JD, Schneider JA, Thoene JG (1981) Adverse reactions to oral cysteamine use in nephropathic cystinosis. Dev Pharmacol Ther 3:25–30CrossRefPubMed
44.
Zurück zum Zitat Besouw MT, Schneider J, Janssen MC, Greco M, Emma F, Cornelissen EA, Desmet K, Skovby F, Nobili F, Lilien MR, De Paepe A, Malfait F, Symoens S, van den Heuvel LP, Levtchenko EN (2013) Copper deficiency in patients with cystinosis with cysteamine toxicity. J Pediatr 163:754–760CrossRefPubMed Besouw MT, Schneider J, Janssen MC, Greco M, Emma F, Cornelissen EA, Desmet K, Skovby F, Nobili F, Lilien MR, De Paepe A, Malfait F, Symoens S, van den Heuvel LP, Levtchenko EN (2013) Copper deficiency in patients with cystinosis with cysteamine toxicity. J Pediatr 163:754–760CrossRefPubMed
45.
Zurück zum Zitat Brodin-Sartorius A, Tete MJ, Niaudet P, Antignac C, Guest G, Ottolenghi C, Charbit M, Moyse D, Legendre C, Lesavre P, Cochat P, Servais A (2012) Cysteamine therapy delays the progression of nephropathic cystinosis in late adolescents and adults. Kidney Int 81:179–189CrossRefPubMed Brodin-Sartorius A, Tete MJ, Niaudet P, Antignac C, Guest G, Ottolenghi C, Charbit M, Moyse D, Legendre C, Lesavre P, Cochat P, Servais A (2012) Cysteamine therapy delays the progression of nephropathic cystinosis in late adolescents and adults. Kidney Int 81:179–189CrossRefPubMed
46.
Zurück zum Zitat Broyer M, Tête M-J (2008) Outcome of cystinosis after 20 years of age. A study of the Enfants-Malades series. Pediatr Nephrol 23:1907CrossRef Broyer M, Tête M-J (2008) Outcome of cystinosis after 20 years of age. A study of the Enfants-Malades series. Pediatr Nephrol 23:1907CrossRef
47.
Zurück zum Zitat Gultekingil Keser A, Topaloglu R, Bilginer Y, Besbas N (2014) Long-term endocrinologic complications of cystinosis. Minerva Pediatr 66:123–130PubMed Gultekingil Keser A, Topaloglu R, Bilginer Y, Besbas N (2014) Long-term endocrinologic complications of cystinosis. Minerva Pediatr 66:123–130PubMed
48.
Zurück zum Zitat Sonies BC, Almajid P, Kleta R, Bernardini I, Gahl WA (2005) Swallowing dysfunction in 101 patients with nephropathic cystinosis: benefit of long-term cysteamine therapy. Medicine (Baltimore) 84:137–146CrossRef Sonies BC, Almajid P, Kleta R, Bernardini I, Gahl WA (2005) Swallowing dysfunction in 101 patients with nephropathic cystinosis: benefit of long-term cysteamine therapy. Medicine (Baltimore) 84:137–146CrossRef
49.
Zurück zum Zitat Tsilou ET, Rubin BI, Reed G, Caruso RC, Iwata F, Balog J, Gahl WA, Kaiser-Kupfer MI (2006) Nephropathic cystinosis: posterior segment manifestations and effects of cysteamine therapy. Ophthalmology 113:1002–1009CrossRefPubMed Tsilou ET, Rubin BI, Reed G, Caruso RC, Iwata F, Balog J, Gahl WA, Kaiser-Kupfer MI (2006) Nephropathic cystinosis: posterior segment manifestations and effects of cysteamine therapy. Ophthalmology 113:1002–1009CrossRefPubMed
50.
Zurück zum Zitat Vaisbich MH, Koch VH (2010) Report of a Brazilian multicenter study on nephropathic cystinosis. Nephron Clin Pract 114:c12–c18CrossRefPubMed Vaisbich MH, Koch VH (2010) Report of a Brazilian multicenter study on nephropathic cystinosis. Nephron Clin Pract 114:c12–c18CrossRefPubMed
52.
Zurück zum Zitat Liang H, Labbé A, Le Mouhaer J, Plisson C, Baudouin C (2017) A new viscous cysteamine eye drops treatment for ophthalmic cystinosis: an open-label randomized comparative phase III pivotal study. Invest Ophthalmol Vis Sci 58:2275–2283CrossRefPubMed Liang H, Labbé A, Le Mouhaer J, Plisson C, Baudouin C (2017) A new viscous cysteamine eye drops treatment for ophthalmic cystinosis: an open-label randomized comparative phase III pivotal study. Invest Ophthalmol Vis Sci 58:2275–2283CrossRefPubMed
Metadaten
Titel
Effects of long-term cysteamine treatment in patients with cystinosis
verfasst von
Gema Ariceta
Vincenzo Giordano
Fernando Santos
Publikationsdatum
19.12.2017
Verlag
Springer Berlin Heidelberg
Erschienen in
Pediatric Nephrology / Ausgabe 4/2019
Print ISSN: 0931-041X
Elektronische ISSN: 1432-198X
DOI
https://doi.org/10.1007/s00467-017-3856-4

Weitere Artikel der Ausgabe 4/2019

Pediatric Nephrology 4/2019 Zur Ausgabe

Update Pädiatrie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.