Skip to main content

A registry of achondroplasia: a 6-year experience from the Czechia and Slovak Republic

Abstract

Background

Achondroplasia (ACH) is one of the most prevalent genetic forms of short-limbed skeletal dysplasia, caused by gain-of-function mutations in the receptor tyrosine kinase FGFR3. In August 2021, the C-type natriuretic peptide (CNP) analog vosoritide was approved for the treatment of ACH. A total of six other inhibitors of FGFR3 signaling are currently undergoing clinical evaluation for ACH. This progress creates an opportunity for children with ACH, who may gain early access to the treatment by entering clinical trials before the closure of their epiphyseal growth plates and cessation of growth. Pathophysiology associated with the ACH, however, demands a long observational period before admission to the interventional trial. Public patient registries can facilitate the process by identification of patients suitable for treatment and collecting the data necessary for the trial entry.

Results

In 2015, we established the prospective ACH registry in the Czechia and the Slovak Republic (http://www.achondroplasia-registry.cz). Patient data is collected through pediatric practitioners and other relevant specialists. After informed consent is given, the data is entered to the online TrialDB system and stored in the Oracle 9i database. The initial cohort included 51 ACH children (average age 8.5 years, range 3 months to 14 years). The frequency of selected neurological, orthopedic, or ORL diagnoses is also recorded. In 2015–2021, a total of 89 measurements of heights, weights, and other parameters were collected. The individual average growth rate was calculated and showed values without exception in the lower decile for the appropriate age. Evidence of paternal age effect was found, with 58.7% of ACH fathers older than the general average paternal age and 43.5% of fathers older by two or more years. One ACH patient had orthopedic limb extension and one patient received growth hormone therapy. Low blood pressure or renal impairment were not found in any patient.

Conclusion

The registry collected the clinical information of 51 pediatric ACH patients during its 6 years of existence, corresponding to ~ 60% of ACH patients living in the Czechia and Slovak Republic. The registry continues to collect ACH patient data with annual frequency to monitor the growth and other parameters in preparation for future therapy.

Background

Achondroplasia (ACH) is one of the most common non-lethal genetic forms of short stature in humans, with an incidence between 1:15,000 to 1:40,000 live births [1,2,3,4]. ACH is caused by activating mutations in FGFR3 receptor tyrosine kinase, which transduces the extracellular communication signals mediated by fibroblast growth factors [5]. The ACH mutations are inherited in an autosomal dominant pattern and localize predominantly to the transmembrane and kinase domains of FGFR3. Four other short stature conditions similar to ACH are also caused by FGFR3 mutations, i.e., hypochondroplasia, SADDAN (severe achondroplasia with developmental delay and acanthosis nigricans) and thanatophoric dysplasia type I and type II. These conditions are collectively termed the FGFR3-related skeletal dysplasias [6]. In ACH, the G380R substitution in the transmembrane domain of FGFR3 accounts for 99% of cases [7] and activates FGFR3 signaling through increased spontaneous dimerization and autophosphorylation [8].

Growth plate cartilage chondrocytes are the primary expression site of FGFR3 in mammals [9]. Aberrant activation of FGFR3 alters several signaling pathways necessary for proper chondrocyte proliferation and differentiation such as WNT, cytokine/STAT, BMP, and Hedgehog signaling [10,11,12,13]. FGFR3 induces chondrocyte proliferation arrest, degradation of the cartilaginous extracellular matrix, and premature senescence, collectively leading to disruption of growth plate architecture and impaired endochondral ossification [14, 15].

ACH is a non-lethal skeletal dysplasia with an average life expectancy decreased by 10 years [16, 17]. The hallmark clinical features of ACH are disproportionate short stature with an adult height of 112–145 cm, midface hypoplasia due to diminished growth of the base of the skull, rhizomelic limb shortening due to growth plate malfunction, and vertebral pedicle shape alterations [18]. Arising complications include spinal stenosis and hydrocephalus, which stem from changes in the foramen magnum and spine [19,20,21]. Sleep apnea is attributed either to upper airway obstruction, related to reduced midface hypoplasia [22,23,24], or myelopathy, related to the cervical spinal cord due to the reduced diameter of the foramen magnum [25,26,27,28]. Sudden death from foramen magnum stenosis was reported and has been substantially reduced by neurosurgical interventions [29, 30]. Heart disease‐related mortality between ages 25 and 35 was over ten times higher than the general population and is attributed to obesity [31, 32]. Other complications include acute and chronic otitis media, which can result in hearing impairment and delay in speech acquisition [33].

The first ACH therapeutic, vosoritide (Voxzogo), was approved in August 2021. Vosoritide is based on a C-type natriuretic peptide (CNP), which is a physiological regulator of bone growth [34]. In clinical trials, vosoritide increased average growth velocity gain over natural growth in ACH patients over a period of up to two years [35, 36]. These results are encouraging although long-term vosoritide efficacy (an absolute gain in stature height) is difficult to estimate since the clinical trials only present evidence in ACH children in growth phases between their 5th and 14th year [35]. In addition to vosoritide, a total of six other inhibitors of FGFR3 signaling are being evaluated in ACH clinical trials [37]. These include CNP variants CNP-PEG and ASB20123, as well as conceptually different inhibitors, which target FGFR3 catalytic activity (infigratinib), inhibit FGFR3 downstream signaling (meclizine), or act as extracellular traps for FGFR3 ligands (recifercept, RBM-007).

Patient age is a factor in admission to the trial, as ACH can only be treated prior to epiphyseal growth plate closure. ACH pathophysiology however demands a long observational period before admission to the interventional trial, limiting the chances to enter the trial for many patients. Clinical registries collecting electronic health records can facilitate access to clinical trials by compiling information on comorbidities, family history, social involvement, and psychological aspects [38]. Because ACH is a rare disease it is estimated that only one out of ten pediatricians will take care of any ACH patient during their lifetime career. Thus the standardization of data reporting and analysis in the registries is also necessary [39]. In this article, we report the establishment of the ACH registry in the Czechia and Slovak Republic, and a six-year experience with running the registry.

Results and discussion

No epidemiological data regarding ACH incidence was available for the Czechia or Slovak Republic before 2015. Based on reported ACH incidence (1/15,000–40,000 live births) [20], we estimate ~ 80 pediatric ACH patients in growth phases under 14 years of age are living in the Czechia and Slovak Republic (combined population 15 million, natality 150,000 children/year). The Registry of Achondroplasia (ReACH; www.achondroplasia-registry.cz) was opened in November 2015. Patient recruitment and reporting were facilitated by educational workshops at the annual meetings of local patient focus groups (ospalecek.cz, paleckovia.sk). The registry gathered a total of 76 patient contacts. The majority of patients had genetically verified ACH, two patients had pseudoachondroplasia, and five patients had hypochondroplasia. Among the 62 pediatric ACH patients were 59 children in growth phase, i.e. aged 14 years or less. A total of 51 patients provided one or more follow-ups (60% of the estimated ACH pediatric population); eight patients were not included in the study due to incomplete data.

All Czech and Slovak ACH patients in the registry carry a G380R substitution in FGFR3, generated by G to A (98% of cases) or G to C (2% of cases) substitution at the FGFR3 nucleotide 1138. All ACH patients were sporadic cases. This is in contrast to the published evidence reporting 5–20% of familial ACH [40]. This may be due to social stigmatization and institutional trust/distrust in affected families [41]. The median paternal age was acquired from the Czech Statistics Institute only for the region of South Moravia, as this data is not available for the whole Czechia territory. Compared with the median paternal age, the mean difference of ACH paternal age was 2.65 ± 6.4 years (average ± S.D., n = 46). There were 58.7% of ACH fathers older than the general average paternal age. The paternal age effect, defined as a father being two or more years older than the general population [42], was present in 43.5% of fathers (Table 1).

Table 1 Paternal age effect in ACH patients

Stature height, body weight, and occipital-frontal circumference measurements were collected totaling 89 individual measurements (52 for 29 boys, 37 for 22 girls). Figure 1 shows stature height values for girls and boys. Repeated height measurements were obtained for 20 patients, enabling calculation of the annualized growth velocity (AGV) in 12 boys and 8 girls (Table 2). Height measurement differences were calculated as interval growth value in centimeters per year and described with annual frequency. On an individual level, AGVs were in the lowest 10% percentile of available AGV growth charts [43].

Fig. 1
figure 1

Growth charts of ACH patients. Body length measurements for girls (A) and boys (B) in centimeters at given age (years). All measurements were below the 3rd percentile of regional growth curves. 97th and 3rd height percentiles are calculated (grey lines)

Table 2 Annualized growth velocity in ACH patients

Among the most frequent comorbidities (Table 3) was acute otitis media (AOM), which affected 24 (47.1%) patients with ~ 50% of AOM diagnoses in the first two years of life (Fig. 2A). The first AOM occurred at 1.5 ± 0.7 years, which is earlier when compared with a large non-ACH AOM cohort showing an average age of first episode at 2.58 years [44]. 83.3% of ACH children experienced more than one AOM (Fig. 2B) in contrast to the general population where recurrent AOM is reported only in about 20.3% of children [44, 45]. These findings correspond with high incidence of chronic otitis media in ACH [45]. Moreover, 66.7% of ACH patients vs. 5.5% of the general population [48] had more than two AOMs, and 45.8% vs. 1% had more than three AOMs. Two ACH patients suffered from 15 and 20 AOMs.

Table 3 Comorbidities in ACH patients
Fig. 2
figure 2

Otitis in ACH patients. Acute otitis media (AOM) affected 24 (47.1%, n = 51) patients, and was diagnosed in the majority of affected individuals in their first 2 years of life (A). Almost all ACH children experienced more than one episode of AOM (20/24; 83.3%), 66.7% were having more than two AOM, and 45.8%. had more than three episodes. Recorded were patients presenting 15 and 20 AOM episodes (B)

The absence of comorbidities such as hypertension or renal failure, which is crucial for C-natriuretic peptide-based therapies for ACH [15, 46,47,48], was not observed in the registry cohort. No arthritis or osteoporosis was recorded. Orthopedic care was described in 49 of 51 patients (96.1%) mostly for tibial malformation, tibial bowing, gibbus, and kyphosis. Spinal stenosis and hydrocephalus were reported in 30 patients (58.8%) (Table 3) and required neurosurgical treatment in four cases. All 30 patients receive continuous neurological care. In 22 (43.1%) patients the parents reported sleep rhonchopathy (snoring) and five patients (9.8%) underwent a sleep study and had confirmed sleep apnea. One patient underwent surgical prolongation of tibial bones during their follow-up. One patient received growth hormone (somatotropin study; Novonordisk, NCT01516229). The evaluation of the quality of life was not carried-out, however, it is of substantial interest [49] and will be collected by the registry in near future.

Conclusions

In summary, the ReACH registry collected clinical information from 51 pediatric ACH patients during its six years of existence, corresponding to ~ 60% of ACH patients aged 3 months to 14 years that were anticipated to live in the Czechia and Slovak Republic. The registry enables the first assessment of the current level of ACH diagnostics and care in both countries.

Methods

The patient data were collected under the Masaryk University and the Office for Personal Data Protection of Czech Republic protocol no. 45/2015, approved by the institutional ethical committee on December 16th, 2015. Patients were approached by genetic counselors, general pediatrics practitioners, or through patient organizations (ospalecek.cz, palcekovia.sk). For contact, a basic online form was prepared and presented at the registry website (http://www.achondroplasia-registry.cz). Patient inclusion criteria were the following: (1) completed informed consent form, (2) clinically diagnosed skeletal dysplasia verified by genetics or orthopedic evaluation, and (3) sufficient data acquisition. For patient evaluation, a complex set of questions and check box answers was prepared as a case report form (CRF; Additional file 1) including necessary epidemiological data but also exclusion criteria for possible clinical studies. Online data collection was based on a TrialDB system (Yale University, Connecticut, USA), where a unique ID is generated for each patient, data transfer is encrypted, and the system is designed to prevent any unauthorized use. Data are stored on the central server at Masaryk University in Brno in the Oracle 9i database. During data recording, a number of pediatric practices preferred the paper version of CRF (Additional file 1), which were exchanged through postal service.

Limitations

As the patient registration is voluntary the registry cohort does not reflect all ACH patients living in the Czechia and Slovak Republic. Several existing ACH families, known to the patient organizations, did not accept invitation to the registry or provide sufficient data and were not included in the study. These families also included familial ACH cases; no familial ACH is presented in the registry. The unequal distribution of care is obvious and results in underdiagnosed comorbidities such as sleep apnea. Leading specialization in ACH care appears to be orthopedic in Czechia [50], and endocrinology in the Slovak Republic. Precise estimation of paternal age is another limitation, as it is available only for one of the 14 existing regions in Czechia.

Availability of data and materials

Please contact the author for data requests.

References

  1. Gardner RJ. A new estimate of the achondroplasia mutation rate. Clin Genet. 1977;11(1):31–8.

    Article  CAS  PubMed  Google Scholar 

  2. Oberklaid F, Danks DM, Jensen F, Stace L, Rosshandler S. Achondroplasia and hypochondroplasia. Comments on frequency, mutation rate, and radiological features in skull and spine. J Med Genet. 1979;16(2):140–6.

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  3. Orioli IM, Castilla EE, Barbosa-Neto JG. The birth prevalence rates for the skeletal dysplasias. J Med Genet. 1986;23(4):328–32.

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  4. Waller DK, Correa A, Vo TM, Wang Y, Hobbs C, Langlois PH, et al. The population-based prevalence of achondroplasia and thanatophoric dysplasia in selected regions of the US. Am J Med Genet A. 2008;146A(18):2385–9.

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  5. Itoh N, Ornitz DM. Fibroblast growth factors: from molecular evolution to roles in development, metabolism and disease. J Biochem (Tokyo). 2011;149(2):121–30.

    Article  CAS  Google Scholar 

  6. Krakow D, Rimoin DL. The skeletal dysplasias. Genet Med. 2010;12(6):327–41.

    Article  PubMed  Google Scholar 

  7. Xue Y, Sun A, Mekikian PB, Martin J, Rimoin DL, Lachman RS, et al. FGFR3 mutation frequency in 324 cases from the International Skeletal Dysplasia Registry. Mol Genet Genomic Med. 2014;2(6):497–503.

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  8. He L, Horton W, Hristova K. Physical basis behind achondroplasia, the most common form of human dwarfism. J Biol Chem. 2010;285(39):30103–14.

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  9. Peters K, Ornitz D, Werner S, Williams L. Unique expression pattern of the FGF receptor 3 gene during mouse organogenesis. Dev Biol. 1993;155(2):423–30.

    Article  CAS  PubMed  Google Scholar 

  10. Li C, Chen L, Iwata T, Kitagawa M, Fu XY, Deng CX. A Lys644Glu substitution in fibroblast growth factor receptor 3 (FGFR3) causes dwarfism in mice by activation of STATs and ink4 cell cycle inhibitors. Hum Mol Genet. 1999;8(1):35–44.

    Article  CAS  PubMed  Google Scholar 

  11. Krejci P, Prochazkova J, Bryja V, Jelinkova P, Pejchalova K, Kozubik A, et al. Fibroblast growth factor inhibits interferon γ-STAT1 and interleukin 6-STAT3 signaling in chondrocytes. Cell Signal. 2009;21(1):151–60.

    Article  CAS  PubMed  Google Scholar 

  12. Qi H, Jin M, Duan Y, Du X, Zhang Y, Ren F, et al. FGFR3 induces degradation of BMP type I receptor to regulate skeletal development. Biochim Biophys Acta. 2014;1843(7):1237–47.

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  13. Buchtova M, Oralova V, Aklian A, Masek J, Vesela I, Ouyang Z, et al. Fibroblast growth factor and canonical WNT/β-catenin signaling cooperate in suppression of chondrocyte differentiation in experimental models of FGFR signaling in cartilage. Biochim Biophys Acta. 2015;1852(5):839–50.

    Article  CAS  PubMed  Google Scholar 

  14. Foldynova-Trantirkova S, Wilcox WR, Krejci P. Sixteen years and counting: the current understanding of fibroblast growth factor receptor 3 (FGFR3) signaling in skeletal dysplasias. Hum Mutat. 2012;33(1):29–41.

    Article  CAS  PubMed  Google Scholar 

  15. Ornitz DM, Legeai-Mallet L. Achondroplasia: development, pathogenesis, and therapy. Dev Dyn Off Publ Am Assoc Anat. 2017;246(4):291–309.

    CAS  Google Scholar 

  16. Wynn J, King TM, Gambello MJ, Waller DK, Hecht JT. Mortality in achondroplasia study: A 42-year follow-up. Am J Med Genet A. 2007;143A(21):2502–11.

    Article  PubMed  Google Scholar 

  17. Hecht JT, Francomano CA, Horton WA, Annegers JF. Mortality in achondroplasia. Am J Hum Genet. 1987;41(3):454–64.

    CAS  PubMed Central  PubMed  Google Scholar 

  18. Srikumaran U, Woodard EJ, Leet AI, Rigamonti D, Sponseller PD, Ain MC. Pedicle and spinal canal parameters of the lower thoracic and lumbar vertebrae in the achondroplast population. Spine. 2007;32(22):2423–31.

    Article  PubMed  Google Scholar 

  19. Horton WA. Dwarfism–an overview Recent developments in diagnostic and genetic concepts. J Kans Med Soc. 1978;79(2):58–61.

    CAS  PubMed  Google Scholar 

  20. Horton WA, Hall JG, Hecht JT. Achondroplasia. Lancet Lond Engl. 2007;370(9582):162–72.

    Article  CAS  Google Scholar 

  21. Wright MJ, Irving MD. Clinical management of achondroplasia. Arch Dis Child. 2012;97(2):129–34.

    Article  CAS  PubMed  Google Scholar 

  22. Waters KA, Everett F, Sillence DO, Fagan ER, Sullivan CE. Treatment of obstructive sleep apnea in achondroplasia: evaluation of sleep, breathing, and somatosensory-evoked potentials. Am J Med Genet. 1995;59(4):460–6.

    Article  CAS  PubMed  Google Scholar 

  23. Zaffanello M, Cantalupo G, Piacentini G, Gasperi E, Nosetti L, Cavarzere P, et al. Sleep disordered breathing in children with achondroplasia. World J Pediatr. 2017;13(1):8–14.

    Article  PubMed  Google Scholar 

  24. Unger S, Bonafé L, Gouze E. Current care and investigational therapies in achondroplasia. Curr Osteoporos Rep. 2017;15(2):53–60.

    Article  PubMed Central  PubMed  Google Scholar 

  25. Mador MJ, Tobin MJ. Apneustic breathing. A characteristic feature of brainstem compression in achondroplasia? Chest. 1990;97(4):877–83.

    Article  CAS  PubMed  Google Scholar 

  26. Onodera K, Niikuni N, Chigono T, Nakajima I, Sakata H, Motizuki H. Sleep disordered breathing in children with achondroplasia. Part 2. Relationship with craniofacial and airway morphology. Int J Pediatr Otorhinolaryngol. 2006;70(3):453–61.

    Article  PubMed  Google Scholar 

  27. Smid CJ, Legare JM, Modaff P, Pauli RM. Apparently benign craniocervical signs in achondroplasia: ‘neurologic leftovers’ identified through a retrospective dataset. Orphanet J Rare Dis. 2020;15(1):301.

    Article  PubMed Central  PubMed  Google Scholar 

  28. Shim Y, Ko JM, Cho TJ, Kim SK, Phi JH. Predictors of cervical myelopathy and hydrocephalus in young children with achondroplasia. Orphanet J Rare Dis. 2021;16(1):81.

    Article  PubMed Central  PubMed  Google Scholar 

  29. Hecht JT, Nelson FW, Butler IJ, Horton WA, Scott CI, Wassman ER, et al. Computerized tomography of the foramen magnum: achondroplastic values compared to normal standards. Am J Med Genet. 1985;20(2):355–60.

    Article  CAS  PubMed  Google Scholar 

  30. Pauli RM, Horton VK, Glinski LP, Reiser CA. Prospective assessment of risks for cervicomedullary-junction compression in infants with achondroplasia. Am J Hum Genet. 1995;56(3):732–44.

    CAS  PubMed Central  PubMed  Google Scholar 

  31. Saint-Laurent C, Garde-Etayo L, Gouze E. Obesity in achondroplasia patients: from evidence to medical monitoring. Orphanet J Rare Dis. 2019;14(1):253.

    Article  PubMed Central  PubMed  Google Scholar 

  32. Hashmi SS, Gamble C, Hoover-Fong J, Alade AY, Pauli RM, Modaff P, et al. Multicenter study of mortality in achondroplasia. Am J Med Genet A. 2018;176(11):2359–2354.

    Article  PubMed  Google Scholar 

  33. Ireland PJ, Johnson S, Donaghey S, Johnston L, McGill J, Zankl A, et al. Developmental milestones in infants and young Australasian children with achondroplasia. J Dev Behav Pediatr. 2010;31(1):41–7.

    Article  PubMed  Google Scholar 

  34. Pejchalova K, Krejci P, Wilcox WR. C-natriuretic peptide: an important regulator of cartilage. Mol Genet Metab. 2007;92(3):210–5.

    Article  CAS  PubMed  Google Scholar 

  35. Savarirayan R, Irving M, Bacino CA, Bostwick B, Charrow J, Cormier-Daire V, et al. C-type natriuretic peptide analogue therapy in children with achondroplasia. N Engl J Med. 2019;381(1):25–35.

    Article  CAS  PubMed  Google Scholar 

  36. Savarirayan R, Tofts L, Irving M, Wilcox W, Bacino CA, Hoover-Fong J, et al. Once-daily, subcutaneous vosoritide therapy in children with achondroplasia: a randomised, double-blind, phase 3, placebo-controlled, multicentre trial. Lancet Lond Engl. 2020;396(10252):684–92.

    Article  CAS  Google Scholar 

  37. Fafilek B, Bosakova M, Krejci P. Expanding horizons of achondroplasia treatment: current options and future developments. Osteoarthritis Cartil. 2021;S1063–4584(21):00980–8.

    Google Scholar 

  38. Cowie MR, Blomster JI, Curtis LH, Duclaux S, Ford I, Fritz F, et al. Electronic health records to facilitate clinical research. Clin Res Cardiol. 2017;106(1):1–9.

    Article  PubMed  Google Scholar 

  39. Kodra Y, Weinbach J, Posada-de-la-Paz M, Coi A, Lemonnier SL, van Enckevort D, et al. Recommendations for improving the quality of rare disease registries. Int J Environ Res Public Health. 2018;15(8):1644.

    Article  PubMed Central  Google Scholar 

  40. Baujat G, Legeai-Mallet L, Finidori G, Cormier-Daire V, Le Merrer M. Achondroplasia. Best Pract Res Clin Rheumatol. 2008;22(1):3–18.

    Article  CAS  PubMed  Google Scholar 

  41. Gollust SE, Apse K, Fuller BP, Miller PS, Biesecker BB. Community involvement in developing policies for genetic testing: assessing the interests and experiences of individuals affected by genetic conditions. Am J Public Health. 2005;95(1):35–41.

    Article  PubMed Central  PubMed  Google Scholar 

  42. Goriely A, Wilkie AOM. Paternal age effect mutations and selfish spermatogonial selection: causes and consequences for human disease. Am J Hum Genet. 2012;90(2):175–200.

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  43. Tanner JM, Davies PS. Clinical longitudinal standards for height and height velocity for North American children. J Pediatr. 1985;107(3):317–29.

    Article  CAS  PubMed  Google Scholar 

  44. Liese JG, Silfverdal SA, Giaquinto C, Carmona A, Larcombe JH, Garcia-Sicilian J, et al. Incidence and clinical presentation of acute otitis media in children aged <6 years in European medical practices. Epidemiol Infect. 2014;142(8):1778–88.

  45. Hunter AG, Bankier A, Rogers JG, Sillence D, Scott CI. Medical complications of achondroplasia: a multicentre patient review. J Med Genet. 1998;35(9):705–12.

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  46. Legeai-Mallet L. C-type natriuretic peptide analog as therapy for achondroplasia. Endocr Dev. 2016;30:98–105.

    Article  CAS  PubMed  Google Scholar 

  47. Igaki T, Itoh H, Suga S, Hama N, Ogawa Y, Komatsu Y, et al. C-type natriuretic peptide in chronic renal failure and its action in humans. Kidney Int Suppl. 1996;55:S144–7.

    CAS  PubMed  Google Scholar 

  48. Caniffi C, Prentki Santos E, Cerniello FM, Tomat AL, González Maglio D, Toblli JE, et al. Cardiac morphological and functional changes induced by C-type natriuretic peptide are different in normotensive and spontaneously hypertensive rats. J Hypertens. 2020;38(11):2305–17.

    Article  CAS  PubMed  Google Scholar 

  49. Witt S, Kolb B, Bloemeke J, Mohnike K, Bullinger M, Quitmann J. Quality of life of children with achondroplasia and their parents - a German cross-sectional study. Orphanet J Rare Dis. 2019;14(1):194.

    Article  PubMed Central  PubMed  Google Scholar 

  50. Zemková D, Krásnicanová H, Marik I. Prediction of the growth in patients with achondroplasia. Arztl Jugendkd. 1991;82(2):237–41.

    Google Scholar 

Download references

Acknowledgements

We thank the physicians and patient families for continuous support and participation in the registry, and to patient organizations Palecek (Czechia) and Paleckovia (Slovak Republic) for disseminating information about the registry among members. We also thank the staff of the Institute of Biostatistics and Analyses of Masaryk University for technical support, to Ing. Karel Adam (Czech Statistics Institute, Brno, Czech Republic) for providing data on paternal age, and to Jennifer Zieba (University of California, Los Angeles) for critical reading of the manuscript.

Funding

Supported by Agency for Healthcare Research of the Czech Republic (NV18-08-00567).

Author information

Authors and Affiliations

Authors

Contributions

MP and PK wrote manuscript. LS and JS contributed to registry design. MP was responsible for patient recruitment and inclusion, and periodic data collection. HV, LS, and JS contributed to data management. JS performed the statistical analysis. HV prepared figures and tables. PK provided funding. All authors read and approved the final manuscript.

Corresponding author

Correspondence to Pavel Krejci.

Ethics declarations

Ethics approval and consent to participate

The study was approved by the Ethical Committee of the Medical Faculty, Masaryk University on December 16th, 2015 as “Registry of Achondroplasia”, and the Office for Personal Data Protection of Czech Republic (protocol no. 45/2015).

Consent for publication

Not applicable.

Competing interests

The authors have no competing interests.

Additional information

Publisher's Note

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

Supplementary Information

Additional file 1

. Case Report Form (CRF) used for reporting the ACH patient data by collaborating pediatricians.

Rights and permissions

Open Access This 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.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Pesl, M., Verescakova, H., Skutkova, L. et al. A registry of achondroplasia: a 6-year experience from the Czechia and Slovak Republic. Orphanet J Rare Dis 17, 229 (2022). https://doi.org/10.1186/s13023-022-02374-x

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s13023-022-02374-x

Keywords