Skip to main content
Erschienen in: Orphanet Journal of Rare Diseases 1/2021

Open Access 01.12.2021 | Research

Phenotypic characterization of X-linked hypophosphatemia in pediatric Spanish population

verfasst von: Enrique Rodríguez-Rubio, Helena Gil-Peña, Sara Chocron, Leire Madariaga, Francisco de la Cerda-Ojeda, Marta Fernández-Fernández, Carmen de Lucas-Collantes, Marta Gil, María Isabel Luis-Yanes, Inés Vergara, Juan David González-Rodríguez, Susana Ferrando, Montserrat Antón-Gamero, Marta Carrasco Hidalgo-Barquero, Angustias Fernández-Escribano, Mº Ángeles Fernández-Maseda, Laura Espinosa, Aniana Oliet, Antonio Vicente, Gema Ariceta, Fernando Santos, RenalTubeGroup

Erschienen in: Orphanet Journal of Rare Diseases | Ausgabe 1/2021

Abstract

Background

X-linked hypophosphatemia (XLH) is a hereditary rare disease caused by loss-of-function mutations in PHEX gene leading tohypophosphatemia and high renal loss of phosphate. Rickets and growth retardation are the major manifestations of XLH in children, but there is a broad phenotypic variability. Few publications have reported large series of patients. Current data on the clinical spectrum of the disease, the correlation with the underlying gene mutations, and the long-term outcome of patients on conventional treatment are needed, particularly because of the recent availability of new specific medications to treat XLH.

Results

The RenalTube database was used to retrospectively analyze 48 Spanish patients (15 men) from 39 different families, ranging from 3 months to 8 years and 2 months of age at the time of diagnosis (median age of 2.0 years), and with XLH confirmed by genetic analysis. Bone deformities, radiological signs of active rickets and growth retardation were the most common findings at diagnosis. Mean (± SEM) height was − 1.89 ± 0.19 SDS and 55% (22/40) of patients had height SDS below—2. All cases had hypophosphatemia, serum phosphate being − 2.81 ± 0.11 SDS. Clinical manifestations and severity of the disease were similar in both genders. No genotype—phenotype correlation was found. Conventional treatment did not attenuate growth retardation after a median follow up of 7.42 years (IQR = 11.26; n = 26 patients) and failed to normalize serum concentrations of phosphate. Eleven patients had mild hyperparathyroidism and 8 patients nephrocalcinosis.

Conclusions

This study shows that growth retardation and rickets were the most prevalent clinical manifestations at diagnosis in a large series of Spanish pediatric patients with XLH confirmed by mutations in the PHEX gene. Traditional treatment with phosphate and vitamin D supplements did not improve height or corrected hypophosphatemia and was associated with a risk of hyperparathyroidism and nephrocalcinosis. The severity of the disease was similar in males and females.
Hinweise
The original online version of this article was updated to amend the article’s funding section.
A correction to this article is available online at https://​doi.​org/​10.​1186/​s13023-021-01786-5.

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Background

X-linked hypophosphatemic rickets (XLH) (OMIM 307800) (ORPHA 89936) is the most common hereditary rickets [15] with an estimated prevalence of 1:20,000 [6, 7]. It follows an X-linked dominant transmission [8]. The disease is caused by a defective function of PHEX gene [1, 913], leading to elevated circulating concentrations of fibroblast growth factor 23 (FGF23) [14], relatively low levels of 1,25 dihydroxyvitamin d [1,25(OH)2D], hyperphosphaturia secondary to decreased proximal tubular reabsorption of phosphate and hypophosphatemia [8, 10, 15]. Classical, conventional treatment of XLH is based on the administration of phosphate supplements and 1-alpha hydroxylated derivates of vitamin D [16]. The wider availability of genetic studies and the recent development of an anti-FGF23 antibody, burosumab, as novel and promising therapy [10, 17] have resulted in a growing current interest for XLH.
We here report the clinical manifestations at diagnosis and follow-up of a large series of Spanish patients included in the online database RenalTube [18]. This study is justified at least by the following reasons: (1) XLH is a rare disease and few publications provide data on large series of patients; (2) XLH has a broad phenotypic variability and additional information is required to better characterize the clinical spectrum of the disease and to explain why the number of cases diagnosed usually does not correspond to the estimated prevalence of the disease; (3) It is important to share data of patients with genetically confirmed XLH in order to facilitate the finding of a potential phenotype—genotype correlation and to have current data that can be compared for the assessment of the new therapies.

Results

Forty-eight patients included in the RenalTube database with the diagnosis of XLH confirmed by defect-of-function variants found in the PHEX gene were analyzed. All variants had been identified as pathogenic. Sixteen patients (33%) had variants with strong evidence of pathogenicity (nonsense, frameshift, deletions) while the other 32 (67%) harbored variants with very strong evidence of pathogenicity (SNPs). Demographic and genetic data from patients are shown in Table 1. Patients were from 39 families and were being followed in pediatric nephrology units of 17 Spanish hospitals (Fig. 1). Fifteen patients were males and 33 females. Median age at diagnosis was 2.0 (IQR 2.6) years and the age ranged from 3 months to 8 years 2 months.
Table 1
Demographic and genetic data of 48 patients belonging to 39 families (Roman number indicates family)
Patient
Relationship
Sex
cDNA mutation
Protein mutation
Variant type
I.1
Index
F
c.758_759delTT
p.F26Cfs
P (PVS1)
I.2
Sister
F
c.758_759delTT
p.F26Cfs
P (PVS1)
II.1
Index
F
c.2223_2224delAC
p.A514Afs516X
P (PVS1)
III.1
Index
F
c.1578_1579delAA
p.K299Nfs304X
P (PVS1)
IV.1
Index
M
c.893A>T
p.N71I
P (PS1)
IV.2
Brother
M
c.893A>T
p.N71I
P (PS1)
V.1
Index
F
c.2633G>C
p.R651P
P (PS1)
VI.1
Index
F
c.1885C>T
p.Q402X
P (PVS1)
VII.1
Index
M
c.?-2664dup2949-?
Splice region variant
P (PVS1)
VII.2
Brother
M
c.?-2664dup2949-?
Splice region variant
P (PVS1)
VII.3
Daughter
F
c.?-2664dup2949-?
Splice region variant
P (PVS1)
VII.4
Daughter
F
c.?-2664dup2949-?
Splice region variant
P (PVS1)
VIII.1
Index
F
c.886insT
p.K69X
P (PVS1)
IX.1
Index
F
c.2048G>A
p.W456X
P (PVS1)
X.1
Index
F
g.22099152G>T
Splice region variant
P (PVS1)
XI.1
Index
M
c.2648_?del
p.A656_?del
P (PVS1)
XII.1
Index
F
c.2327_?del
p.R549_?del
P (PVS1)
XIII.1
Index
F
g.22168393_delA
Splice region variant
P (PVS1)
XIV.1
Index
F
c.1552C>T
p.R291X
P (PVS1)
XV.1
Index
F
g.22190503G>A
Splice region variant
P (PVS1)
XVI.1
Index
F
c.889_893delGTAAA
p.V70Sfs77X
P (PVS1)
XVII.1
Index
F
c.2086_?del
p.A469_?del
P (PVS1)
XVIII.1
Index
F
c.1180T>C
p.W167R
P (PS1)
XIX.1
Index
F
c.2282C>T
p.P534L
P (PS1)
XX.1
Index
F
c.2416G>A
p.G579R
P (PS1)
XXI.1
Index
F
c.2920C>T
p.R747X
P (PVS1)
XXII.1
Index
F
c.2920C>T
p.R747X
P (PVS1)
XXIII.1
Index
M
c.1152delA
p.L157Lfs220X
P (PVS1)
XXIV.1
Index
F
c.1572C>A,c.1580_1582delTGA
p.Y297X
P (PVS1)
XXV.1
Index
F
g.22076478A>T
Splice region variant
P (PVS1)
XXVI.1
Index
F
g.22190507G>A
Splice region variant
P (PVS1)
XXVII.1
Index
F
c.889_893delGTAAA
p.V70Sfs77X
P (PVS1)
XXVII.2
Father
M
c.889_893delGTAAA
p.V70Sfs77X
P (PVS1)
XXVIII.1
Index
M
c.2879G>T
p.C733F
P (PS1)
XXVIII.2
Sister
F
c.2879G>T
p.C733F
P (PS1)
XXIX.1
Index
F
c.2642T>C
p.F654S
P (PS1)
XXX.1
Index
M
c.2387T>G
p.L569R
P (PS1)
XXX.2
Mother
F
c.2387T>G
p.L569R
P (PS1)
XXXI.1
Index
F
c.2085G>C
p.K468N
P (PS1)
XXXII.1
Index
M
c.2282C>T
p.P534L
P (PS1)
XXXIII.1
Index
M
g.22033125T>G
Splice region variant
P (PVS1)
XXXIV.1
Index
M
c.2380C>T
p.R567X
P (PVS1)
XXXV.1
Index
M
c.2005G>T
p.V442P
P (PS1)
XXXV.2
Mother
F
c.2005G>T
p.V442P
P (PS1)
XXXVI.1
Index
F
c.2416G>A
G579R
P (PS1)
XXXVII.1
Index
M
g.22099152G>A
Splice region variant
P (PVS1)
XXXVIII.1
Index
F
c.2617delG
p.D646Ifs
P (PVS1)
XXXIX.1
Index
M
c.1363_1364delTC
p.S228Pfs236X
P (PVS1)
F: female, M: male
P: pathogenic
PVS1: very strong evidence of pathogenicity according to reference 19
PS1: strong evidence of pathogenicity according to reference 19
Presenting manifestations are shown in Table 2 for each patient. Bone deformities and radiological signs of active rickets were the most frequent findings leading to diagnosis. Ten patients were diagnosed because of family screening. Age at diagnosis of these patients was no different from that of the rest of the series as it ranged from 0.5 to 8 years with a median age of 1.04 years.
Table 2
Clinical manifestations at diagnosis
Patient
Age at diagnosis
Bone deformities
Active rickets
Longitudinal growth retardation (≤ 2 SDS)
Dental problems
I.1
3 m
No
No
I.2
1 y
Yes
Yes
II.1
1 y 5 m
Yes
Yes
Yes
No
III.1
1 y 6 m
Yes
Yes
Yes
IV.1
1 y 4 m
No
No
Yes
Yes
IV.2
4 y
No
No
No
No
V.1
5 y
Yes
Yes
Yes
No
VI.1
2 y
Yes
No
No
No
VII.1
8 y
VII.2
2 y
Yes
No
VII.3
11 m
Yes
Yes
No
VII.4
1 y
Yes
VIII.1
4 y
Yes
Yes
No
No
IX.1
5 y
Yes
Yes
No
No
X.1
5 y
Yes
Yes
Yes
No
XI.1
2 y 3 m
Yes
Yes
Yes
No
XII.1
2 y
Yes
Yes
XIII.1
5 y
Yes
XIV.1
9 m
Yes
Yes
No
No
XV.1
1 y 1 m
Yes
Yes
Yes
XVI.1
4 y
Yes
Yes
No
XVII.1
5 y
Yes
XVIII.1
2 y
Yes
Yes
Yes
No
XIX.1
7 m
Yes
Yes
No
XX.1
2 y 1 m
Yes
Yes
XXI.1
4 y 5 m
Yes
Yes
No
No
XXII.1
4 y
Yes
Yes
Yes
No
XIII.1
2 y
Yes
Yes
No
No
XXIV.1
3 y
Yes
No
XXV.1
1 y 6
Yes
Yes
No
No
XXVI.1
4 y
Yes
Yes
Yes
No
XXVII.1
6 m
Yes
Yes
No
No
XXVII.2
8 y
Yes
No
XXVIII.1
1 y 6 m
Yes
No
XXVIII.2
6 m
Yes
Yes
No
No
XXIX.1
1 y 9 m
Yes
Yes
Yes
No
XXX.1
1 y 1 m
Yes
Yes
Yes
XXX.2
1 y 6 m
Yes
No
XXXI.1
8 y2 m
Yes
Yes
Yes
No
XXXII.1
1 y 6 m
Yes
Yes
No
No
XXXIII.1
5 y
Yes
XXXIV.1
2 y 2 m
Yes
Yes
No
No
XXXV.1
7 m
Yes
Yes
No
XXXV.2
2 y 6 m
Yes
Yes
Yes
XXXVI.1
6 m
Yes
Yes
No
No
XXXVII.1
1 y 9 m
Yes
Yes
Yes
Yes
XXXVIII.1
1 y 6 m
Yes
Yes
No
No
XXXIX.1
2 y 2 m
Yes
Yes
No
Percentage of patients
P/A/U
P/A/U
P/A/U
P/A/U
73/6/21
73/6/21
46/38/17
6/63/31
m: month, y: year
SDS: standard deviation score. Dash: information in this field was missing from the database
P/A/U: present/absent/unreported
Twenty-two out of40 patients (55%) in whom the height was registered presented growth retardation (height ≤ 2 SDS). Patients’ height (X ± SEM) was − 1.89 ± 0.19 SDS (n = 40) (Fig. 2). In 87% (35/40) the height was below the 50th percentile. Weight was − 0.88 ± 0.14 SDS (n = 41) and body mass index 0.2 ± 0.15 SDS (n = 40).
Biochemical findings at diagnosis are shown in Table 3. Mean values (± SEM) of available data were serum phosphate 2.7 ± 0.1 mg/dl; − 2.81 ± 0.11 SDS, (n = 41), alkaline phosphatase (892 ± 84 mU/ml) (n = 39), 1,25(OH)2D62 ± 7 pg/ml (n = 34), parathyroid hormone (PTH)70 ± 7 pg/ml (n = 33), and tubular phosphate reabsorption (TPR) 69 ± 4% (n = 26).
Table 3
Biochemical manifestations at diagnosis
Patient
Serum phosphate
Serum alkaline phosphatases (mU/ml)
Serum 1,25(OH)2D (pg/ml)
Serum intact PTH (pg/ml)
TPR (%)
mg/dl
SDS
I.1
3.4
− 2.21
1230
87
I.2
3.8
− 1.05
1916
76
II.1
2.6
− 2.68
695
57
81
79
III.1
2.1
− 3.36
1646
16
68
32
IV.1
2.8
− 2.41
378
73
29
78
IV.2
3.0
− 2.44
232
40
43
82
V.1
2.1
− 3.99
1513
147
78
85
VI.1
2.6
− 2.68
525
33
64
78
VII.1
VII.2
2.4
− 2.95
516
VII.3
991
28
30
VII.4
VIII.1
3.1
− 2.27
598
55
39
38
IX.1
3.0
− 2.44
1824
47
59
67
X.1
2.9
− 2.61
639
33
136
74
XI.1
2.3
− 3.09
58
XII.1
2.7
− 2.54
48
XIII.1
XIV.1
3.3
− 2.34
892
64
65
75
XV.1
2.0
− 3.49
704
20
39
XVI.1
2.4
− 3.47
571
108
71
26
XVII.1
2.5
− 3.30
28
30
XVIII.1
2.3
− 3.09
1864
31
64
73
XIX.1
XX.1
2.9
− 2.27
446
88
116
93
XXI.1
3.1
− 2.27
470
56
49
86
XXII.1
2.8
− 2.78
697
74
32
76
XXIII.1
2.1
− 3.36
733
68
82
XXIV.1
2.2
− 3.22
514
61
78
58
XXV.1
2.9
− 2.27
1940
40
57
58
XXVI.1
2.3
− 3.64
423
82
XXVII.1
2.9
− 2.86
432
31
23
XXVII.2
XXVIII.1
3.0
− 2.14
236
78
54
XXVIII.2
3.3
− 2.34
426
171
57
90
XXIX.1
1.8
− 3.77
1555
XXX.1
3.1
− 2.00
829
183
101
88
XXX.2
2.2
− 3.22
158
25
XXXI.1
XXXII.1
2.8
− 2.41
692
41
111
XXXIII.1
2.1
− 3.99
76
54
XXXIV.1
2.3
− 3.09
856
64
101
46
XXXV.1
3.5
− 2.08
916
227
82
XXXV.2
2.2
− 3.22
1620
70
XXXVI.1
2.8
− 2.41
1093
65
66
XXXVII.1
2.4
− 2.95
1527
46
94
XXXVIII.1
2.4
− 2.95
856
40
58
XXXIX.1
3.0
− 2.14
759
51
43
1,25(OH)2D: 1,25 dihydroxyvitamin D. PTH: Parathyroid hormone
TPR: tubular phosphate reabsorption. Dash: information in this field was missing from the database
No differences were found between males and females for clinical manifestations, growth impairment or biochemical data at diagnosis. Likewise, no genotype–phenotype correlation was found. Actually, even patients within the same family presented different severity of clinical and biochemical manifestations.
Growth and biochemical variables of 26 patients after a median follow uptime of 7.42 years (IQR = 11.26) are shown in Table 4, Figs. 3 and 4. Anthropometric data were − 1.94 ± 0.16 SDS for height (n = 24), − 0.82 ± 0.10 SDS for weight (n = 22) and 0.14 ± 0.19 SDS for BMI (n = 22). Comparison of data from patients with information both at diagnosis and last follow-up showed mean variations of 0.13 ± 0.23 SDS for height (p > 0.05) (n = 20), 0.35 ± 0.14 SDS for weight (p = 0.02) (n = 20) and 0.13 ± 0.20 SDS for BMI (p > 0.05) (n = 20).
Table 4
Biochemical manifestations at last follow up
Patient
Follow-up Time
Serum phosphate
Serum alkaline phosphatases (mU/ml)
Serum 1,25(OH)2D
(pg/ml)
Serum intact PTH (pg/ml)
TPR (%)
mg/dl
SD (Z score)
I.1
7 y 2 m
2.9
− 2.07
787
30
72
I.2
10 y 1 m
2.7
− 2.90
1035
35
50
II.1
1 y 1 m
3.8
− 1.05
34
70
III.1
IV.1
7 y 6 m
3.0
− 1.91
231
78
32
60
IV.2
7 y 6 m
2.6
− 3.06
266
73
44
48
V.1
VI.1
VII.1
VII.2
VII.3
8 y 4 m
1.9
− 3.69
77
42
VII.4
8 y 3 m
1.8
− 3.85
665
108
73
VIII.1
2 y 7 m
2.4
− 3.47
868
15
84
81
IX.1
X.1
7 y 5 m
3.0
− 2.41
962
50
93
85
XI.1
5 y 11 m
2.8
− 2.78
632
45
49
42
XII.1
11 y 5 m
2.8
− 2.23
636
112
86
XIII.1
17 y 6 m
1.7
− 4.22
29
86
64
XIV.1
2 y 1 m
2.8
− 2.41
96
20
70
XV.1
25 y 5 m
2.0
− 3.55
20
75
45
XVI.1
XVII.1
18 y 8 m
1.7
− 4.22
120
32
99
42
XVIII.1
XIX.1
26 y 2 m
2.4
− 2.66
200
XX.1
1 y 9 m
3.3
− 1.73
312
72
53
45
XXI.1
3 y 5 m
2.6
− 2.56
348
43
26
53
XXII.1
XXIII.1
2 y 10 m
3.0
− 2.44
29
28
XXIV.1
XXV.1
16 y
1.9
− 3.77
37
XXVI.1
2 y 4 m
2.8
− 2.78
240
86
25
70
XXVII.1
1 y 10 m
2.8
− 2.41
58
49
76
XXVII.2
37 y 4 m
2.3
− 2.88
117
80
XXVIII.1
XXVIII.2
XXIX.1
1 y 10 m
4.8
0.31
244
68
73
89
XXX.1
XXX.2
XXXI.1
XXXII.1
XXXIII.1
4 y
2.1
− 3.36
114
76
XXXIV.1
XXXV.1
XXXV.2
XXXVI.1
XXXVII.1
XXXVIII.1
XXXIX.1
1,25(OH)2D: 1,25 dihydroxyvitamin D
m: month, y: year
PTH: parathyroid hormone
TPR: tubular phosphate reabsorption. Dash: information in this field was missing from the database
Mean SDS for serum phosphate was − 2.72 ± 0.20 (n = 25). Alkaline phosphatases, 1,25 (OH)2D and PTH levels were 525 ± 82 mU/ml (n = 14), 53 ± 7 pg/ml (n = 15) and 68 ± 8 pg/ml (n = 26) respectively. Mean tubular phosphate reabsorption was 65 ± 3% (n = 22). Comparison between diagnosis and last follow-up data revealed a variation of 0.20 ± 0.28 SDS for serum phosphate (p > 0.05) (n = 20), 5 ± 7 pg/ml for 1,25 (OH)2D (p > 0.05) (n = 10), 0 ± 11 pg/ml for PTH (p > 0.05) (n = 16) and − 7 ± 8 for tubular phosphate reabsorption (p > 0.05) (n = 11).
Eight out of 24 patients with renal ultrasounds at last follow-up presented nephrocalcinosis (Table 5).
Table 5
Treatment and clinical data at last follow-up
Patient
Phosphorus element dose (mg/kg/day)
Vitamin D dose (µg/day)
Nephrocalcinosis
I.1
38a
0.50c
No
I.2
40a
0.50c
No
II.1
86a
1.20d
Yes
III.1
IV.1
83a
0.60d
No
IV.2
62a
1.00d
No
V.1
VI.1
VII.1
No
VII.2
VII.3
55b
0.50c
Yes
VII.4
52b
0.50c
Yes
VIII.1
90a
1.50d
Yes
IX.1
X.1
40a
0.25d
No
XI.1
83a
0.50c
No
XII.1
44b
0.29d
No
XIII.1
41b
0.25c
Yes
XIV.1
52a
1.20d
XV.1
41b
1.00c
No
XVI.1
XVII.1
45b
0.25c
Yes
XVIII.1
XIX.1
27b
0.50c
Yes
XX.1
32a
0.50c
No
XXI.1
63a
1.50d
No
XXII.1
XXIII.1
65a
1.10d
No
XXIV.1
XXV.1
38a
1.40c
XXVI.1
58b
0.25c
No
XXVII.1
48b
0.60d
No
XXVII.2
XXVIII.1
XXVIII.2
XXIX.1
0.10d
No
XXX.1
XXX.2
Yes
XXXI.1
XXXII.1
XXXIII.1
49b
0.75c
No
XXXIV.1
XXXV.1
XXXV.2
XXXVI.1
XXXVII.1
XXXVIII.1
XXXIX.1
aPhosphate was administered as a solution
bPhosphate was administered as tablets
cCorresponds to 1,25 dihydroxy vitamin D
dCorresponds to 1 hydroxy vitamin D

Discussion

This study provides a current description of the phenotypic characteristics of a large cohort of Caucasian pediatric patients with XLH genetically confirmed. The sample is a broad representation of the Spanish children with XLH, coming from several hospitals scattered through the country and provides data at diagnosis and after a median follow-up of 7.42 years. The study confirms that growth retardation, bone deformities and active lesions of rickets are the main presenting manifestations of the disease, within a wide spectrum of symptoms. No significant differences were found between males and females as for the severity of the disease. It is of interest that a broad spectrum of PHEX gene variants, all of them already described as pathogenic, was found and no mutation was specifically prevalent in Spanish population. All variants were classified as pathogenic according to the American College of Medical Genetics and Genomics consensus [19]. There was a high phenotypical variability even among family members harboring the same mutations, suggesting that other genes and environmental factors may affect the severity of XLH, as reported by other authors [20, 21].
In addition, the study shows that conventional treatment with phosphate supplements and vitamin D metabolites does not lead to persistent correction of hypophosphatemia or reduction of renal wasting of phosphate and does not modify the circulating levels of calcitriol. Unfortunately, this study does not provide information on circulating FGF23 levels, given its retrospective design. At the last follow-up visit, 11 out of 25 patients had serum PTH values mildly elevated. In XLH, development of hyperparathyroidism is thought to be related with the pharmacological administration of phosphate [22]. Eight patients developed nephrocalcinosis during the follow-up period, a finding linked to the administration of phosphate and vitamin D that usually does not result in subsequent clinical complications [16].
Though conventional treatment has been described to heal active signs of rickets and may improve bone deformities [23], this study confirms that it does not lead to catch-up growth. Mean height Z score of the group of patients remained low, − 1.89 at diagnosis versus − 1.94 at the last visit, although Fig. 3 indicates that the individual patients’ response varied from marked improvement to worsening of growth impairment. Two patients, VII.3 and XV.1, transiently received growth hormone treatment and their heights improved + 2.19and + 0.66 SDS, respectively. It is of note that 4 out of 16 patients had BMI greater than + 1.00 SDS at the last follow-up visit. This percentage of 25% corresponds to the normal distribution of reference population and it indicates that tendency to overweight and obesity was not found in the group of XLH patients here reported, unlike other series that have recently drawn attention to these complications likely related with the sedentary life and restricted mobility of these patients [11]. In this regard, a slight but significant increase in weight was found during the follow-up period in our series.
Our study presents methodological limitations inherent to the retrospective analysis and to the fact that patients’ information was extracted from a database in which some data were missing and cannot be recovered. It is also of note the lack of information on the degree of adherence to medication of each patient as well as the different monitoring protocols among the participating centers. However, it is an observational clinical study describing a large cohort of Spanish pediatric patients with genetically confirmed XLH and it provides current and interesting information on the clinical and biochemical features of the disease, at diagnosis and follow-up after conventional treatment. Our findings could be used as reference for further studies using burosumab treatment.

Conclusions

This study confirms that growth retardation and rickets were the most prevalent clinical manifestations at diagnosis in a large series of Spanish pediatric patients with XLH confirmed by identification of pathogenic variants in the PHEX gene. Traditional treatment with phosphate supplements and calcitriol did not improve height or corrected hypophosphatemia and was associated with a risk of hyperparathyroidism and nephrocalcinosis. The severity of the disease was similar in males and females and no phenotype-genotype association was found.

Patients and methods

The RenalTube database including 48 patients, 15 males and 33 females, with the diagnosis of XLH confirmed by defect-of-function mutations found in the PHEX gene was retrospectively reviewed to obtain demographic information and clinical and biochemical manifestations at diagnosis and at the last annual follow-up. Genetic information was confirmed and formatted according to Genome Reference Consortium Human Build 38 patch release 13 (GRCh38.p13) [24]. Variants were analyzed in silico and classified according to recommendations from the consensus of the American College of Medical Genetics and Genomics and the Association for Molecular Pathology [19] as pathogenic, likely pathogenic, benign or likely benign. Results for the age are presented as median and interquartile range (IQR). Other variables are presented for the group as mean (X) ± standard error of the mean (SEM). Z score (SDS) of anthropometric values was calculated using Spanish age and sex-matched reference values [25]. Patients with height ≤ 2 SDS were considered to have longitudinal growth retardation according to World Health Organization standards [26]. Reference values for biochemical parameters were obtained from the laboratory of the Hospital Universitario Central de Asturias (HUCA) [27].
All patients received treatment with phosphate supplementation (dose range of phosphorus element: 27–90 mg/kg/day at last follow-up) and vitamin D metabolites (dose range: 0.25–1.5 µg/day at last follow-up), according to the criteria and indications given by their physicians (Table 5). Two patients (VII.3 and XV.1) received growth hormone treatment. None of them received burosumab treatment.
Information in RenalTube database was downloaded and formatted to an Excel database. All fields but reasons for consultation and genetic information were multichoice or numeric format.
Chi squared test was used to analyze differences between sex for binary (Yes/No) fields (growth retardation, bone deformities). F-test was used to assess variance equality between sex for anthropometric and biochemical values. T-test was used to analyze differences between sex for anthropometric and biochemical values. Paired T-test for means was used to analyze differences between diagnosis and last follow-up for anthropometric and biochemical values. T-test for unequal variances was used to compare age at diagnosis for patients with and without family history of the disease.
Phenotype—genotype correlation was assessed by isolating the most severe phenotypes (lowest serum concentrations, most severe growth retardation, highest levels of alkaline phosphatases) and comparing genetic mutations in these patients looking for big deletions, SNPs with entirely different amino acids or nonsense mutations.

Acknowledgements

Not applicable.
Patients information has been processed via RenalTube database where patients signed their consent to participate in scientific studies.
Patients or parents signed consent for publication of their data through RenalTube consent form.

Competing interests

The study has been partially funded by Kyowa Kirin Farmacéutica S.L.U. This company produces the drug Crysvita® (burosumab). Nevertheless, no patients in this study had been or were being treated with burosumab at the time of data collection.
Open AccessThis article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://​creativecommons.​org/​licenses/​by/​4.​0/​. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Literatur
1.
Zurück zum Zitat Bastepe M, Jüppner H. Inherited hypophosphatemic disorders in children and the evolving mechanisms of phosphate regulation. Rev Endocr Metab Disord. 2008;9(2):171–80.CrossRef Bastepe M, Jüppner H. Inherited hypophosphatemic disorders in children and the evolving mechanisms of phosphate regulation. Rev Endocr Metab Disord. 2008;9(2):171–80.CrossRef
2.
Zurück zum Zitat Fuente R, Gil-Peña H, Claramunt-Taberner D, Hernández-Frías O, Fernández-Iglesias Á, Alonso-Durán L, et al. MAPK inhibition and growth hormone: a promising therapy in XLH. FASEB J. 2019;33(7):8349–62.CrossRef Fuente R, Gil-Peña H, Claramunt-Taberner D, Hernández-Frías O, Fernández-Iglesias Á, Alonso-Durán L, et al. MAPK inhibition and growth hormone: a promising therapy in XLH. FASEB J. 2019;33(7):8349–62.CrossRef
3.
Zurück zum Zitat Morey M, Castro-Feijóo L, Barreiro J, Cabanas P, Pombo M, Gil M, et al. Genetic diagnosis of X-linked dominant hypophosphatemic rickets in a cohort study: Tubular reabsorption of phosphate and 1,25(OH)2D serum levels are associated with PHEX mutation type. BMC Med Genet. 2011;12(1):116.CrossRef Morey M, Castro-Feijóo L, Barreiro J, Cabanas P, Pombo M, Gil M, et al. Genetic diagnosis of X-linked dominant hypophosphatemic rickets in a cohort study: Tubular reabsorption of phosphate and 1,25(OH)2D serum levels are associated with PHEX mutation type. BMC Med Genet. 2011;12(1):116.CrossRef
6.
Zurück zum Zitat Francis F, Hennig S, Korn B, Reinhardt R, de Jong P, Poustka A, et al. A gene (PEX) with homologies to endopeptidases is mutated in patients with X–linked hypophosphatemic rickets. Nat Genet. 1995;11(2):130–6.CrossRef Francis F, Hennig S, Korn B, Reinhardt R, de Jong P, Poustka A, et al. A gene (PEX) with homologies to endopeptidases is mutated in patients with X–linked hypophosphatemic rickets. Nat Genet. 1995;11(2):130–6.CrossRef
8.
Zurück zum Zitat Gattineni J, Baum M. Genetic disorders of phosphate regulation. Pediatr Nephrol. 2012;27(9):1477–87.CrossRef Gattineni J, Baum M. Genetic disorders of phosphate regulation. Pediatr Nephrol. 2012;27(9):1477–87.CrossRef
10.
Zurück zum Zitat Carpenter TO, Whyte MP, Imel EA, Boot AM, Högler W, Linglart A, et al. Burosumab therapy in children with X-linked hypophosphatemia. N Engl J Med. 2018;378(21):1987–98.CrossRef Carpenter TO, Whyte MP, Imel EA, Boot AM, Högler W, Linglart A, et al. Burosumab therapy in children with X-linked hypophosphatemia. N Engl J Med. 2018;378(21):1987–98.CrossRef
11.
Zurück zum Zitat Zhukouskaya VV, Rothenbuhler A, Colao A, Di SC, Kamenický P, Trabado S, et al. Increased prevalence of overweight and obesity in children with x-linked hypophosphatemia. Endocr Connect. 2020;9(2):144–53.CrossRef Zhukouskaya VV, Rothenbuhler A, Colao A, Di SC, Kamenický P, Trabado S, et al. Increased prevalence of overweight and obesity in children with x-linked hypophosphatemia. Endocr Connect. 2020;9(2):144–53.CrossRef
12.
Zurück zum Zitat Emma F, Cappa M, Antoniazzi F, Bianchi ML, Chiodini I, Eller Vainicher C, et al. X-linked hypophosphatemic rickets: An Italian experts’ opinion survey. Ital J Pediatr. 2019;45(1):1–7.CrossRef Emma F, Cappa M, Antoniazzi F, Bianchi ML, Chiodini I, Eller Vainicher C, et al. X-linked hypophosphatemic rickets: An Italian experts’ opinion survey. Ital J Pediatr. 2019;45(1):1–7.CrossRef
13.
Zurück zum Zitat Cho HY, Lee BH, Kang JH, Ha IS, Cheong HI, Choi Y. A clinical and molecular genetic study of hypophosphatemic rickets in children. Pediatr Res. 2005;58(2):329–33.CrossRef Cho HY, Lee BH, Kang JH, Ha IS, Cheong HI, Choi Y. A clinical and molecular genetic study of hypophosphatemic rickets in children. Pediatr Res. 2005;58(2):329–33.CrossRef
14.
Zurück zum Zitat Endo I, Fukumoto S, Ozono K, Namba N, Inoue D, Okazaki R, et al. Nationwide survey of fibroblast growth factor 23 (FGF23)-related hypophosphatemic diseases in Japan: prevalence, biochemical data and treatment. Endocr J. 2015;62(9):811–6.CrossRef Endo I, Fukumoto S, Ozono K, Namba N, Inoue D, Okazaki R, et al. Nationwide survey of fibroblast growth factor 23 (FGF23)-related hypophosphatemic diseases in Japan: prevalence, biochemical data and treatment. Endocr J. 2015;62(9):811–6.CrossRef
15.
Zurück zum Zitat Ariceta G, Langman CB. Growth in X-linked hypophosphatemic rickets. Eur J Pediatr. 2007;166(4):303–9.CrossRef Ariceta G, Langman CB. Growth in X-linked hypophosphatemic rickets. Eur J Pediatr. 2007;166(4):303–9.CrossRef
16.
Zurück zum Zitat Haffner D, Emma F, Eastwood DM, Duplan MB, Bacchetta J, Schnabel D, et al. Clinical practice recommendations for the diagnosis and management of X-linked hypophosphataemia. Nat Rev Nephrol. 2019;15:435–55.CrossRef Haffner D, Emma F, Eastwood DM, Duplan MB, Bacchetta J, Schnabel D, et al. Clinical practice recommendations for the diagnosis and management of X-linked hypophosphataemia. Nat Rev Nephrol. 2019;15:435–55.CrossRef
17.
Zurück zum Zitat Insogna KL, Briot K, Imel EA, Kamenický P, Ruppe MD, Portale AA, et al. A randomized, double-blind, placebo-controlled, phase 3 trial evaluating the efficacy of burosumab, an anti-FGF23 antibody, in adults with X-linked hypophosphatemia: week 24 primary analysis. J Bone Miner Res. 2018;33(8):1383–93.CrossRef Insogna KL, Briot K, Imel EA, Kamenický P, Ruppe MD, Portale AA, et al. A randomized, double-blind, placebo-controlled, phase 3 trial evaluating the efficacy of burosumab, an anti-FGF23 antibody, in adults with X-linked hypophosphatemia: week 24 primary analysis. J Bone Miner Res. 2018;33(8):1383–93.CrossRef
19.
Zurück zum Zitat Richards S, Aziz N, Bale S, Bick D, Das S, Gastier-Foster J, Grody WW, Hegde M, Lyon E, Spector E, Voelkerding KR. Standards and guidelines for the interpretation of sequence variants: a joint consensus recommendation of the american college of medical genetics and genomics and the association for molecular pathology. Genet Med. 2015;17(5):405–24.CrossRef Richards S, Aziz N, Bale S, Bick D, Das S, Gastier-Foster J, Grody WW, Hegde M, Lyon E, Spector E, Voelkerding KR. Standards and guidelines for the interpretation of sequence variants: a joint consensus recommendation of the american college of medical genetics and genomics and the association for molecular pathology. Genet Med. 2015;17(5):405–24.CrossRef
20.
Zurück zum Zitat Holm IA, Nelson AE, Robinson BG, Mason RS, Marsh DJ, Cowell CT, et al. Mutational analysis and genotype-phenotype correlation of the PHEX gene in X-linked hypophosphatemic rickets. J Clin Endocrinol Metab. 2001;86(8):3889–99.CrossRef Holm IA, Nelson AE, Robinson BG, Mason RS, Marsh DJ, Cowell CT, et al. Mutational analysis and genotype-phenotype correlation of the PHEX gene in X-linked hypophosphatemic rickets. J Clin Endocrinol Metab. 2001;86(8):3889–99.CrossRef
21.
Zurück zum Zitat Rafaelsen S, Johansson S, Ræder H, Bjerknes R. Hereditary hypophosphatemia in Norway: a retrospective population-based study of genotypes, phenotypes, and treatment complications. Eur J Endocrinol. 2016;174(2):125–36.CrossRef Rafaelsen S, Johansson S, Ræder H, Bjerknes R. Hereditary hypophosphatemia in Norway: a retrospective population-based study of genotypes, phenotypes, and treatment complications. Eur J Endocrinol. 2016;174(2):125–36.CrossRef
23.
Zurück zum Zitat Carpenter TO, Imel EA, Holm IA, et al. A clinician’s guide to X-linked hypophosphatemia. J Bone Min Res. 2011;26(7):1381–8.CrossRef Carpenter TO, Imel EA, Holm IA, et al. A clinician’s guide to X-linked hypophosphatemia. J Bone Min Res. 2011;26(7):1381–8.CrossRef
27.
Zurück zum Zitat Servicio de Bioquímica Clínica (AGC-Laboratorio de Medicina). Hospital Universitario Central de Asturias. Valores bioquímica HUCA. Biblioteca De Pruebas Bioquimicas. 2013. Servicio de Bioquímica Clínica (AGC-Laboratorio de Medicina). Hospital Universitario Central de Asturias. Valores bioquímica HUCA. Biblioteca De Pruebas Bioquimicas. 2013.
Metadaten
Titel
Phenotypic characterization of X-linked hypophosphatemia in pediatric Spanish population
verfasst von
Enrique Rodríguez-Rubio
Helena Gil-Peña
Sara Chocron
Leire Madariaga
Francisco de la Cerda-Ojeda
Marta Fernández-Fernández
Carmen de Lucas-Collantes
Marta Gil
María Isabel Luis-Yanes
Inés Vergara
Juan David González-Rodríguez
Susana Ferrando
Montserrat Antón-Gamero
Marta Carrasco Hidalgo-Barquero
Angustias Fernández-Escribano
Mº Ángeles Fernández-Maseda
Laura Espinosa
Aniana Oliet
Antonio Vicente
Gema Ariceta
Fernando Santos
RenalTubeGroup
Publikationsdatum
01.12.2021
Verlag
BioMed Central
Erschienen in
Orphanet Journal of Rare Diseases / Ausgabe 1/2021
Elektronische ISSN: 1750-1172
DOI
https://doi.org/10.1186/s13023-021-01729-0

Weitere Artikel der Ausgabe 1/2021

Orphanet Journal of Rare Diseases 1/2021 Zur Ausgabe