You are on page 1of 6

Bone 103 (2017) 144–149

Contents lists available at ScienceDirect

Bone

journal homepage: www.elsevier.com/locate/bone

Full Length Article

Treatment with neridronate in children and adolescents with


osteogenesis imperfecta: Data from open-label, not controlled,
three-year Italian study☆
L. Idolazzi a,⁎, A. Fassio a, O. Viapiana a, M. Rossini a, G. Adami a, F. Bertoldo b, F. Antoniazzi c, D. Gatti a
a
Rheumatology Unit, Department of Medicine, University of Verona, Italy
b
Internal Medicine Unit, Department of Medicine, University of Verona, Italy
c
Pediatric Clinic, Department of Surgical Sciences, Dentistry, Gynecology and Pediatrics, Pediatric Division, University of Verona, Verona, Italy

a r t i c l e i n f o a b s t r a c t

Article history: Introduction: The present study assessed the long-term efficacy and safety of intravenous (i.v.) neridronate in
Received 30 December 2016 children and adolescents affected by osteogenesis imperfecta (OI).
Revised 30 June 2017 Methods: 55 young patients (mean age 12.6 ± 3.9 years) affected by OI were included in the study. Neridronate
Accepted 1 July 2017 was administered by i.v. infusion at a dose of 2 mg/kg (maximum dose of 100 mg) at intervals of three-months
Available online 3 July 2017
for three years. Dual X-ray absorptiometry of the lumbar spine, hip and ultradistal and proximal radius were eval-
uated every 6 months. Blood calcium, phosphate, albumin, fasting urinary calcium/creatinine ratio were obtained
Keywords:
Neridronate
at baseline and every 3 months. Serum bone turnover markers total and bone alkaline phosphatase were per-
Osteogenesis imperfecta formed every 12 months in a proportion of patients.
Safety Results: Mean lumbar spine and total hip bone mineral density (BMD) and bone mineral content significantly in-
Tolerability creased from baseline compared to all subsequent time points (p b 0.001). Mean ultradistal radius BMD signifi-
cantly increased from month 18 (p = 0.026). Levels of bone turnover markers significantly decreased from
baseline to all post-baseline observation time points. There was no statistically significant effect on fracture
risk (p = 0.185), although a significant reduction was observed in the mean number of fractures occurring during
treatment compared to pre-treatment values. The most frequent adverse events were arthralgia, fever, joint
sprain. An acute phase reaction was reported in 26 (22.8%) patients. None of the reported serious adverse events
was considered as treatment-related.
Conclusion: Long-term i.v. neridronate treatment has positive effects on BMD, bone turnover markers and frac-
ture risk with a good safety profile.
© 2017 Elsevier Inc. All rights reserved.

1. Introduction The bone histomorphometric picture is usually characterized by the


presence of an excessive number of osteoblasts with impaired activity
Osteogenesis imperfecta (OI) is a fairly common heritable disorder and consequent defective bone matrix deposition. The trabecular skele-
(one in 15–20,000 births) and represents a generalized connective tis- tal turnover is generally increased, at least up to maturity [5]. These
sue disease. OI is mainly characterized by bone fragility and substantial histomorphometric features have been confirmed by the detection of
growth deficiency [1–3]. Other relevant findings are often non-skeletal an increase in bone turnover markers, such as serum osteocalcin, colla-
features, including blue sclerae, hearing loss, dentinogenesis imperfecta, gen peptide cross-links (N-telopeptide) and calcium urinary excretion
impaired pulmonary function and cardiac valvular regurgitation. Differ- [6–10]. Several therapies have been suggested for the treatment of OI,
ent types of OI have been identified, based on clinical and hereditarily including bisphosphonates [11–15], denosumab [4], growth hormone
characteristics and on biochemical defects [1–4]. alone or in combination with other treatments [16,17], teriparatide
(only in adults) [18,19] and bone marrow transplantation [20]. The in-
☆ Summary: This study evaluated the efficacy and safety of neridronate administered crease in bone turnover markers is a hallmark of the disease and repre-
intravenously (2 mg/kg efficacy, tri-monthly) to children/adolescents with osteogenesis sents the rational for therapy with anti-resorptive agents such as
imperfecta for a period of three years. Neridronate was effective and well tolerated,
bisphosphonates or denosumab [21,22]. To date, bisphosphonates
resulting in a significant increase in bone mineral density, bone mineral content and
reduced number of fractures. have shown to increase bone mineral density (BMD). A positive effect
⁎ Corresponding author at: p.le L Scuro 2, 37100 Verona, Italy. on prevention of fractures both in adults and in children has been re-
E-mail address: luca.idolazzi@univr.it (L. Idolazzi). ported in some studies [15,22,23], but data are generally inconsistent.

http://dx.doi.org/10.1016/j.bone.2017.07.004
8756-3282/© 2017 Elsevier Inc. All rights reserved.
L. Idolazzi et al. / Bone 103 (2017) 144–149 145

Recent systematic reviews have also summarized evidence on use of skeletal sites ranged from 1% to 2.5% in subjects, with a wide range in
bisphosphonates in OI [22,24,25] and they agree that the present evi- bone mass values.
dence is currently insufficient to conclude that bisphosphonate therapy All fractures that occurred over the three years prior to treatment
can improve outcomes other than BMD. It is likely that additional stud- and during the study were validated by X-ray film or radiology reports
ies are still needed since previous trials were underpowered to assess and regularly collected during the study.
outcomes other than BMD and generally had short treatment periods. Serum calcium, phosphate, albumin and fasting urinary calcium/cre-
Regarding bisphosphonates, large placebo-controlled long-term trials atinine ratio were obtained at baseline and every three months. Total al-
in OI are unlikely to be conducted due to several issues such as lack of kaline phosphatase (ALP: Roche Diagnostics) was measured by an
funding or the difficulty of finding patients who are willing to risk immunoassay analyzer and serum bone alkaline phosphatase (BAP,
being randomized to placebo for a long period. Alkphase–B; Metra Biosystems) by an enzyme-linked
Neridronate is an amino-bisphosphonate structurally similar to immunoabsorbent assay. All biochemical measurements were per-
alendronate and pamidronate, differing only in the number of methyl formed every 12 months only in a small proportion of patients. Inter-
groups of the side chain: five for neridronate, three for alendronate, and intra-assay variations ranged from 7% to 12%.
and two for pamidronate [26]. It is available in both intramuscular and Unfortunately, the frequent collection of urine samples in the major-
intravenous formulations, which altogether can prevent a number of ity of patients was not possible and therefore urinary free-deoxy
complications caused by oral bisphosphonates. Neridronate has been pyridinoline analysis was not undertaken.
extensively investigated in patients with OI in Italy [18] where it has
been licensed for the treatment of OI (it is currently the only drug regis- 2.4. Safety
tered for this indication), Complex Regional Pain Syndrome type 1 and
Paget's disease of bone. Adverse events (AEs) were categorized by System Organ Class (SOC)
The aim of this study was to assess the efficacy and safety of and Preferred Term (PT) by using the MedDRA dictionary (version
neridronate, when administered to children and adolescents with OI 11.0). The occurrence of clinical AEs and biochemical alterations (hema-
by intravenous infusions at a dose of 2 mg/kg (up to a maximum of tology, blood chemistry, and urinalysis) were recorded at every visit.
100 mg) once every three months for a period of three years.
2.5. Statistical analysis
2. Methods
Statistical analysis was performed on data obtained from the safety
2.1. Study population population, defined as all patients who received at least one dose of
study medication, and the intention-to-treat population (ITT), defined
The study sample includes patients aged b 20 years and with a defi- as all patients who received at least one dose of study medication and
nite diagnosis of OI, enrolled by a single investigational study team. Pa- with post-baseline data. These two populations were considered in
tients were enrolled from a dedicated outpatient clinic of a National the present study.
Centre of Reference for Osteogenesis Imperfecta. All patients were pre- Demographic and baseline characteristics were summarized by
viously diagnosed on the basis of a history of fragility fractures (one or means of descriptive statistics, and expressed as the number of observa-
more, especially during youth), concomitant positive family history tions, mean, standard deviation, minimum and maximum for continu-
and/or extra skeletal manifestation specific for the disease such as: ous variables, and frequency distributions (number and percentages)
hearing loss, blue sclerae or dentinogenesis imperfecta. Exclusion for categorical variables. Medical history, concomitant diseases (coded
criteria were: previous treatment with bisphosphonates, serious con- using the MedDRA dictionary version 11.0), and previous and concom-
comitant diseases including cardiovascular, hematological, psychiatric itant medication (coded with WHO Drug Dictionary) were collected.
or pulmonary diseases, renal insufficiency with serum creatinine The percent change from baseline of DXA parameters evaluated at
N1.5 mg/dl, history of hypersensitivity to bisphosphonates. Prior to un- the different body sites were analyzed by means of descriptive statistics
dertaking this study, authorization of the Italian ‘Istituto Superiore di and the mean values of the percent changes from baseline were com-
Sanità, the Italian Ministry of Health and the reference Independent pared with 0 by means of a one-sample Student t-test. In order to take
Ethics Committee (IEC) for the participating center was obtained. All pa- account of multiple tests, Bonferroni's correction was applied to the
tients (or a parent) signed informed consent. All procedures performed probability associated with each test.
in the study involving human participants were in accordance with the The percent change from baseline of bone markers parameters was
ethical standards of the institutional and/or national research commit- also analyzed for the densitometric efficacy parameters.
tee and the 1964 Helsinki declaration and its later amendments or com- The frequency of fractures was recorded and the definition of fre-
parable ethical standards. quency of fracture was considered as the number of patients with
new fractures per year observed during the treatment period or record-
ed in the three years prior to commencement of the trial. The number of
2.2. Treatment administered fractures per year before and during the study was also considered
(Table 5). The distribution of rates of patients with fractures observed
Neridronate was administered by i.v. infusion at a dosage of 2 mg/kg, before and during the treatment period was analyzed by the McNemar
up to a maximum of 100 mg in saline solution (0.9% NaCl) (approxi- test. Furthermore, the distribution in the number of patients' fractures
mately 10 mg/100 ml), with an infusion time of about two hours. Infu- in the two periods were compared using the Wilcoxon signed-rank
sions were undertaken every three months. Supplementation with test. All statistical analysis was performed using SPSS version 11 (SPSS
calcium (1000 mg daily) and vitamin D (800 IU daily) was also admin- Inc., Chicago, IL, USA) and a p-value of b0.05 was considered statistically
istered to the patient. significant.

2.3. Efficacy variables 3. Results

Dual X-ray absorptiometry (DXA) of the lumbar spine, hip and The study population comprised 55 patients (mean age 12.6 ±
ultradistal and proximal radius were evaluated every 6 months using 3.9 years), mainly affected by type 1 OI (type 1 OI; 65.4%, type 3 OI;
a full-body bone densitometer (Hologic 4500, Waltham, US). The preci- 18.2%, or type 4 OI; 16.4%). Demographic characteristics are reported
sion error for BMD, BMC (bone mineral content) and area at different in Table 1. The population included a similar proportion of female and
146 L. Idolazzi et al. / Bone 103 (2017) 144–149

Table 1 Analysis of the distribution of the number of patients with or with-


Demographic features and type of OI in the study population. out fractures and their rate during the three years of treatment or in
Characteristic (N = 55) the three years prior to treatment showed that a significant proportion
Gender, N (%)
of patients (38.2%) had at least one fracture before the study and did not
Males, N (%) 30 (54.5) have fractures during the treatment period (p b 0.001 in the distribution
Females, N (%) 25 (45.5) of patients with or without fractures) (Table 5). When the number of
Age, years (mean ± SD) 12.6 ± 3.92 (range 5–19) fractures was taken in account, the mean number of fractures observed
Height, cm (mean ± SD) 131.2 ± 22.85 (range 90–176)
in the three years of treatment was also found to be significantly lower
Weight, kg (mean ± SD) 34.7 ± 14.78 (range 11–70)
Type of OI, N (%) than that observed in the three years before treatment (Wilcoxon
1 36 (65.4%) signed-rank test; p b 0.001) (Table 5).
3 10 (18.2%) The most frequent AEs observed were pyrexia (23.6%), back pain
4 9 (16.4%) (12.7%) and arthralgia (10.9%). An acute phase reaction with flu-like ill-
N = number of patients. ness was reported in 26 (47.3%) patients and was the most frequent AE,
OI = osteogenesis imperfecta. mainly related to the drug. No fatal events occurred. Serious adverse
events (SAEs) were reported in 19 (34.5%) patients. None of the report-
ed SAEs were considered treatment-related. No cases of osteonecrosis of
the jaw (ONJ) or atypical femur fracture were reported.
male patients (45.5% vs. 55.5%) and pathological fractures were report- Laboratory tests (blood count, general blood chemistry and urinaly-
ed in all (N = 55) patients. sis) did not show substantial changes in the results from baseline up to
A total of 51 (92.7%) patients completed the scheduled 36-month 36 months. Clinically significant and non-treatment-related abnormali-
treatment period, while 4 (7.3%) of them discontinued the study early. ties in laboratory parameters were reported in a very small number of
The patients received 11.1 ± 2.49 infusions (minimum 3, maximum patients (N = 3): one with thrombocytopenia, one with AST elevation,
13). A summary of patients who completed or discontinued the study and one with ALT elevation.
and the different reasons for their withdrawal is presented in Table 2.
Spine DXA was only performed in 43 of the 55 (78.2%) patients for 4. Discussion
the presence of dimorphism or more of 2 vertebral fractures involving
the lumbar spine. DXA at the hip was performed in 30 of 55 (54.5%) pa- The present study showed that i.v. neridronate treatment was asso-
tients for the presence of fractures of both femurs. ciated with a marked and statistically significant increase in both BMD
The mean lumbar spine BMD significantly increased from baseline to and BMC at the lumbar spine and total hip from baseline up to
all follow-up time points with percentage changes that progressively in- 36 months of the study (Table 3). Hip DXA is not currently considered
creased from month 6 (+15%) to month 36 (+51%) (Table 3). The in- a diagnostic tool in the pediatric age, but the significant and relevant
crease in BMD was also associated with a concomitant increase in changes documented (also in this skeletal site) are, in our opinion,
BMC and in Area (Table 3). None of the patients with repeated measure- one important effect of neridronate treatment in this severe form of os-
ments of lumbar spine BMD had any lumbar vertebral fractures for the teoporosis. The increase in BMD (about 40–50% after 36 months of ther-
duration of the study. apy) is likely caused by suppression of bone resorption and reduction of
Mean total hip BMD significantly increased from baseline at all sub- bone remodeling as direct consequence [27].
sequent time points, with percentage changes that progressively in- The concomitant increase in bone Area has also important conse-
creased from month 6 (+ 11%) to month 36 (+ 41%) (Table 3). The quences. DXA measures areal BMD and this increase in projected Area
increase in BMD was also associated with a concomitant increase in produces an underestimation of the gain in BMD. On the other hand,
BMC and in Area (Table 3). the increase in bone Area confirms that this anti-reabsorptive drug
The mean lumbar spine and total hip area increased from baseline to would not appear to exert negative effects on skeletal growth [28].
all further time points, except for month 6 for the total hip (Table 3). Neridronate is an amino-bisphosphonate with proven efficacy since
As expected, mean ALP and BAP levels significantly decreased from 2003, when the positive results of a randomized trial involving 46 adults
baseline to all post-baseline time points by about 50% (Table 4). with OI treated for 24 months were published [23]. Two years later data
In the three years before treatment was undertaken, 43 of the 55 pa- from a RCT with similar design on pediatric patients was published [15]
tients (78.2%) incurred at least one fracture. Over the three year study demonstrating that neridronate significantly increases BMD and lowers
period the rate of patients with new fractures decreased to 24/55 the risk of clinical fracture in prepubescent children with OI. These two
(43.6%). positive experiences led to the registration of neridronate for osteogen-
All the fractures taken into account were fragility fractures (sponta- esis imperfecta in Italy for both children and adults [15,23]. Actually,
neous or due to minimal trauma) and the most relevant involved: hip (7 neridronate is also licensed in Italy for the treatment of Paget's disease
patients), shinbone (5 patients), thoracic vertebrae (1 patients), radius of bone and for CRSP1 [18,29]. Bisphosphonate therapy has been used
(1 patient), humerus (1 patient) and pelvis (1 patent). to treat patients with OI for the past three decades [30] and is currently
the most widely available medical treatment for OI, even though many
different compounds, dosage, and protocols have been proposed. The
positive effects of bisphosphonates on areal BMD (by DXA) at the
Table 2
Reasons for study withdrawal.
spine and other skeletal sites are widely recognised [4]. More controver-
sial is the question of what clinically relevant benefit children with OI
Number (%) may have from higher areal BMD due to the fact that some clinically rel-
Patients who completed the study 51 (92.7) evant outcomes such as fracture rate, mobility, and quality of life are dif-
Patients who discontinued the study 4 (7.3) ficult to assess in OI [22,24,25,31].
Reasons for discontinuation
The reported densitometric results appear to be in line with ones fol-
Adverse event 0 (0.0)
Serious adverse events 1 (1.8) lowing other i.v. bisphosphonate therapies such as pamidronate and
Exclusion criteria 0 (0.0) zoledronate [22,24,25]. However, to the best of our knowledge, most
Patient compliance 1 (1.8) of these studies currently available concerning bisphosphonates in OI
Consent withdrawal 2 (3.6) were of shorter duration (12–24 months) than ours (36 months). In-
Economic issues 0 (0.0)
deed, this is one of the strengths of our study, showing that the positive
L. Idolazzi et al. / Bone 103 (2017) 144–149 147

Table 3
Percentage variation in DXA parameters from baseline values.

Month BMD (g/cm2) mean ± SD (95% CI) BMC (g) mean ± SD (95% CI) Area (cm2) mean ± SD (95% CI)

Lumbar spine
6 14.8 ± 11.1 (11.4–18.2)⁎⁎ 31.3 ± 26.4 (23.2–39.5)⁎⁎ 14.7 ± 24.2 (7.2–22.1)⁎
12 26.1 ± 20.3 (19.7–32.4)⁎⁎ 41.1 ± 42.3 (27.9–54.3)⁎⁎ 13.5 ± 35.5 (2.4–24.5)
18 32.8 ± 24.3 (24.7–40.9)⁎⁎ 58.7 ± 46.9 (43.1–74.4)⁎⁎ 19.4 ± 29.7 (9.5–29.3)⁎
24 40.3 ± 28.2 (31.4–49.2)⁎⁎ 69.6 ± 53.7 (52.6–86.5)⁎⁎ 19.9 ± 26.9 (11.5–28.4)⁎⁎
30 43.3 ± 30.7 (33.7–52.9)⁎⁎ 75.3 ± 58.0 (57.2–93.4)⁎⁎ 20.8 ± 25.5 (12.9–28.8)⁎⁎
36 50.7 ± 33.1 (40.0–61.4)⁎⁎ 90.8 ± 68.7 (68.5–113.1)⁎⁎ 24.9 ± 30.1 (15.2–34.7)⁎⁎

Total hip
6 11.3 ± 11.0 (7.3–15.5) 13.1 ± 16.2 (6.8–19.4)⁎ 2.1 ± 8.6 (−1.3–5.4)
12 22.4 ± 20.7 (14.5–30.3)⁎⁎ 33.8 ± 31.6 (21.8–45.8)⁎⁎ 8.7 ± 13.7 (3.5–13.9)⁎
18 33.6 ± 26.7 (22.6–44.6)⁎⁎ 56.0 ± 39.5 (39.7–72.3)⁎⁎ 16.2 ± 16.2 (9.5–22.9)⁎
24 34.1 ± 27.8 (23.7–44.5)⁎⁎ 54.1 ± 44.0 (37.7–70.6)⁎⁎ 13.8 ± 17.3 (7.4–20.3)⁎
30 37.0 ± 29.6 (26.1–47.9)⁎⁎ 60.9 ± 51.3 (41.7–80.1)⁎⁎ 16.5 ± 19.4 (9.3–23.7)⁎⁎
36 41.4 ± 31.4 (29.4–53.4)⁎⁎ 66.1 ± 53.9 (45.2–87.0)⁎⁎ 16.5 ± 22.9 (7.6–25.3)⁎

BMD = bone mineral density. BMC = bone mineral content.


⁎ p b 0.05 vs. baseline.
⁎⁎ p b 0.001 vs. baseline.

densitometric gain of bisphophonates also increases in the third year of considered different from other bone diseases such as osteoporosis: in
treatment, with a protective effect on fracture risk. A recent study has fact in OI the fractures mostly involve the femoral diaphysis and this
now analyzed data from children with OI treated with pamidronate pattern seems to be changed by bisphosphonate treatment [35].
up to 4 years [32]. This study showed an increase in LS BMD for all OI Regardless, during the three years of the study, we did not observe
types and also confirmed a reduction in the incidence of fractures. any drug-related SAE. Furthermore, no cases of atypical fracture or
Placebo-controlled studies should provide high quality data but ONJ were observed, though we should keep in mind that these two
there are issues related to the lack of funding and also to the likely reluc- events are extremely rare and hard to be seen in a study with this sam-
tance of patients to risk to being treated with placebo while a licensed ple size.
pharmacological treatment is already available. For these reasons it The most common side effects were related to the acute phase re-
may be necessary, meanwhile, to obtain clinical data from observational sponse, that sometimes follows aminobisphosphonate administration,
studies. Ideally, these studies should aim to have a long treatment peri- especially if given intravenously and in young patients [36]. These
od and should provide a comparison between the rate of fractures ob- symptoms are not serious, transient and usually appear only after the
served before and during treatment. very first infusion. They never led to treatment discontinuation during
The main strength of the present study is the finding of a statistically this study for any patient. Patients that exited the study did it mainly be-
significant effect on fracture risk. The hallmark of OI are the fragility cause of practical reasons which prevented them from being able to at-
fractures and these patients have an increased fracture rate throughout tend regular visits. This trial was monocentric and as a consequence of
their life. In this study, the percentage of patients with fractures was al- this design many patients had to traveled from distant regions, often
most halved during treatment compared to the three years prior to with difficulties in attending visits.
treatment (43/55 vs. 24/55 respectively) and a significant rate of pa- The results of laboratory tests (hematology, blood chemistry and uri-
tients (37%) who experienced at least one fracture in the three years be- nalysis) did not show substantial changes from baseline up to 36 months
fore the study, had no fracture during the treatment period (p b 0.001 in of any parameters. In particular, there were no alterations in hepatic or
the distribution of patients with or without fractures). This protective renal function.
effect was also confirmed when the number of fracture was taken in The i.v. route for administering bisphosphonates guarantees a com-
account. plete adherence and probably the best bioavailability and it prevents
Besides efficacy, what is considered essential for a treatment is safety
and tolerability.
Bisphosphonates have a good safety profile with a low incidence of
Table 5
serious adverse events such as atypical femur fracture or ONJ. In OI, Number of patients with fractures in the 3 years prior to treatment compared with 3 years
the risk of ONJ induced by bisphosphonates seems to be very low [33, of treatment.
34]. Furthermore, the pattern of femoral fracture in OI should be
Number of patients with fractures

Number (%) of patients with During treatment (3 years)


fractures
Before treatment (3 years) Patients without Patients with
Table 4 fractures, N (%) fractures, N (%)
Percent changes of bone markers considering baseline values. Patients without fractures, N 10 (18.2) 2 (3.6)
= 12 (21.8)
Alkaline phosphatase Bone alkaline phosphatase Patients with fractures, N = 21 (38.2) 22 (9.4)
43 (78.2)
Baseline value 239,6 ± 108,5 (U/L) 100,4 ± 39,4 (mcg/L)
McNemar's test p-value b0.001
Mean (95% CI) N Mean (95% CI) N
Mean number of fractures
Month 12 −25.9 (−31.0 to −20.5)⁎⁎ 48 −28.0 (−36.6 to −18.3)⁎⁎ 57
Month 24 −41.9 (−47.4 to −35.9)⁎⁎ 48 −36.7 (−46.3 to −25.4)⁎⁎ 47 Number of fractures, mean ± SD (range)
Month 36 −49.1 (−56.2 to −40.8)⁎⁎ 44 −52.2 (−64.6 to −35.3)⁎ 24 Before treatment (3 years) 2.7 ± 2.37 (0–8)
During treatment (3 years) 0.9 ± 1.43 (0–7)
N = number of patients.
Wilcoxon signed-rank test p-value b0.001
⁎ p b 0.01 vs. baseline.
⁎⁎ p b 0.001 vs. baseline. N = number of patients.
148 L. Idolazzi et al. / Bone 103 (2017) 144–149

the well-known gastrointestinal side effects responsible for a significant [5] R. Baron, J.M. Gertner, R. Lang, A. Vignery, Increased bone turnover with decreased
bone formation by osteoblasts in children with osteogenesis imperfecta tarda, Pediatr.
rate of treatment discontinuation with bisphosphonates [36]. Res. 17 (1983) 204–207, http://dx.doi.org/10.1203/00006450-198303000-00007.
Our study does have some limitations, such as the sample size and [6] V. Braga, D. Gatti, M. Rossini, et al., Bone turnover markers in patients with osteogen-
lack of a control group. However, we obtained clinically important re- esis imperfecta, Bone 34 (2004) 1013–1016, http://dx.doi.org/10.1016/j.bone.2004.02.
023.
sults on the anti-fracture effects of this treatment. Furthermore, in this [7] C. Cepollaro, S. Gonnelli, C. Pondrelli, et al., Osteogenesis imperfecta: bone
study, a repeated evaluation on spine plan RX was not scheduled and turnover, bone density, and ultrasound parameters, Calcif. Tissue Int. 65
thus it was not possible to look for the typical vertebral reshaping effect (1999) 129–132.
[8] S. Minisola, R. Rosso, E. Romagnoli, Bone resorption assessed by immunoassay of
following bisphosphonate therapy. Another limitation of the study is
urinary cross-linked collagen peptides in patients with osteogenesis imperfecta, J.
the change of pubertal status, possibly involving some of the patients Bone Miner. Res. Off. J. Am. Soc. Bone Miner. Res. 10 (1995) 335–339, http://dx.
during the three-year follow-up. This feature could be a possible con- doi.org/10.1002/jbmr.5650100222.
[9] R.E. Brenner, U. Vetter, A.M. Bollen, et al., Bone resorption assessed by immunoassay
founding factor in our analysis, particularly with regard to densitomet-
of urinary cross-linked collagen peptides in patients with osteogenesis imperfecta, J.
ric change. Bone Miner. Res. Off. J. Am. Soc. Bone Miner. Res. 9 (1994) 993–997, http://dx.doi.
org/10.1002/jbmr.5650090706.
[10] J. Iwamoto, T. Takeda, S. Ichimura, Increased bone resorption with decreased
5. Conclusion activity and increased recruitment of osteoblasts in osteogenesis imperfecta
type I, J. Bone Miner. Metab. 20 (2002) 174–179, http://dx.doi.org/10.1007/
In conclusion, i.v. administration neridronate in OI children and ad- s007740200025.
[11] T. Palomo, M.C. Andrade, B.S.E. Peters, et al., Evaluation of a modified pamidronate
olescents over a three-year period was shown to be effective, safe and protocol for the treatment of osteogenesis imperfecta, Calcif. Tissue Int. 98 (2016)
well tolerated. This study demonstrated a positive effect in terms of 42–48, http://dx.doi.org/10.1007/s00223-015-0061-y.
fracture risk expressed as the number of fractures occurring in patients [12] G.A. Otaify, M.S. Aglan, M.M. Ibrahim, et al., Zoledronic acid in children with osteo-
genesis imperfecta and Bruck syndrome: a 2-year prospective observational study,
in the three years of treatment compared to the three years prior to
Osteoporos. Int. 27 (2016) 81–92 J Establ Result Coop Eur Found Osteoporos Natl
treatment and confirmed the positive densitometric effects of bisphos- Osteoporos Found USA. (doi: 10.1007/s00198-015-3216-9).
phonate therapy that appears to be important for obtaining increased [13] A. Biggin, L. Zheng, J.N. Briody, et al., The long-term effects of switching from active
intravenous bisphosphonate treatment to low-dose maintenance therapy in chil-
peak bone mass.
dren with osteogenesis imperfecta, Horm. Res. Paediatr. 83 (2015) 183–189,
http://dx.doi.org/10.1159/000369582.
Funding [14] G. Eghbali-Fatourechi, Bisphosphonate therapy in pediatric patients, J. Diabetes
Metab. Disord. 13 (2014) 109, http://dx.doi.org/10.1186/s40200-014-0109-y.
[15] D. Gatti, F. Antoniazzi, R. Prizzi, et al., Intravenous neridronate in children with
This research did not receive any specific grant from funding agen- osteogenesis imperfecta: a randomized controlled study, J. Bone Miner. Res. Off.
cies in the public, commercial, or not-for-profit sectors. J. Am. Soc. Bone Miner. Res. 20 (2005) 758–763, http://dx.doi.org/10.1359/
JBMR.041232.
[16] J.C. Marini, S. Bordenick, G. Heavner, et al., The growth hormone and somatomedin
Conflict of interest axis in short children with osteogenesis imperfecta, J. Clin. Endocrinol. Metab. 76
(1993) 251–256, http://dx.doi.org/10.1210/jcem.76.1.8421094.
[17] F. Antoniazzi, E. Monti, G. Venturi, et al., GH in combination with bisphosphonate
Ombretta Viapiana has received speaking fees from Abiogen, Amgen, treatment in osteogenesis imperfecta, Eur. J. Endocrinol. 163 (2010) 479–487 Eur
Merck Sharp & Dohme Corp. Luca Idolazzi has received speaking fees Fed Endocr Soc. (doi: 10.1530/EJE-10-0208).
from Eli Lilly, UCB, Abbvie, Novartis. Angelo Fassio declares no conflict [18] D. Gatti, M. Rossini, O. Viapiana, et al., Clinical development of neridronate: potential
for new applications, Ther. Clin. Risk Manag. 9 (2013) 139–147, http://dx.doi.org/10.
of interest. Francesco Bertoldo reports personal fees from Abiogen, per- 2147/TCRM.S35788.
sonal fees from Amgen, personal fees from Eli Lilly, personal fees from [19] E.S. Orwoll, J. Shapiro, S. Veith, et al., Evaluation of teriparatide treatment in adults
Bayer, personal fees from Italfarmaco, outside the submitted work. with osteogenesis imperfecta, J. Clin. Invest. 124 (2014) 491–498, http://dx.doi.
org/10.1172/JCI71101.
Maurizio Rossini has received speaking and consulting fees from [20] E.M. Horwitz, D.J. Prockop, P.L. Gordon, et al., Clinical responses to bone marrow
Abiogen, Amgen, Eli-Lilly and Merck Sharp & Dohme Corp. Franco transplantation in children with severe osteogenesis imperfecta, Blood 97 (2001)
Antoniazzi declares no conflict of interest. 1227–1231.
[21] H. Hoyer-Kuhn, O. Semler, E. Schoenau, Effect of denosumab on the growing skele-
Giovanni Adami declares no conflict of interest. Davide Gatti has re- ton in osteogenesis imperfecta, J. Clin. Endocrinol. Metab. 99 (2014) 3954–3955,
ceived speaking fees from Abiogen, Amgen, Eli-Lilly and Neopharmed- http://dx.doi.org/10.1210/jc.2014-3072.
Gentili. [22] K. Dwan, C.A. Phillipi, R.D. Steiner, D. Basel, Bisphosphonate therapy for osteogenesis
imperfecta, Cochrane Database Syst. Rev. 7 (2014), CD005088. http://dx.doi.org/10.
1002/14651858.CD005088.pub3.
Acknowledgements [23] S. Adami, D. Gatti, F. Colapietro, et al., Intravenous neridronate in adults with osteo-
genesis imperfecta, J. Bone Miner. Res. Off. J. Am. Soc. Bone Miner. Res. 18 (2003)
126–130, http://dx.doi.org/10.1359/jbmr.2003.18.1.126.
The Authors would like to kindly thank Abiogen Pharma (Pisa) for [24] J.D. Hald, E. Evangelou, B.L. Langdahl, S.H. Ralston, Bisphosphonates for the pre-
providing neridronate (Nerixia®) for the treatment of OI patients for vention of fractures in osteogenesis imperfecta: meta-analysis of placebo-con-
the entire study duration. The Authors would also like to thank Dr. trolled trials, J. Bone Miner. Res. 30 (2015) 929–933, http://dx.doi.org/10.
1002/jbmr.2410.
Fraccarollo Caterina for the ELISA assays, Dr. Poli Fabio for technical as- [25] E.B.G. Rijks, B.C. Bongers, M.J.G. Vlemmix, et al., Efficacy and safety of bisphosphonate
sistance and the LURM (Laboratorio Universitario di Ricerca Medica) therapy in children with osteogenesis imperfecta: a systematic review, Horm. Res.
Research Center, University of Verona where the biochemical analyses Paediatr. 84 (2015) 26–42, http://dx.doi.org/10.1159/000381713.
[26] H. Fleisch, Bisphosphonates: mechanisms of action, Endocr. Rev. 19 (1998) 80–100,
were performed. http://dx.doi.org/10.1210/edrv.19.1.0325.
[27] S. Adami, J.A. Kanis, Assessment of involutional bone loss: methodological and concep-
References tual problems, J. Bone Miner. Res. Off. J. Am. Soc. Bone Miner. Res. 10 (1995) 511–517,
http://dx.doi.org/10.1002/jbmr.5650100402.
[1] A. Forlino, J.C. Marini, Osteogenesis imperfecta, Lancet 387 (2016) 1657–1671 Lond [28] F.H. Glorieux, Treatment of osteogenesis imperfecta: who, why, what? Horm. Res.
Engl. (doi: 10.1016/S0140-6736(15)00728-X). 68 (Suppl. 5) (2007) 8–11, http://dx.doi.org/10.1159/000110463.
[2] A. Forlino, W.A. Cabral, A.M. Barnes, J.C. Marini, New perspectives on osteogenesis [29] D. Gatti, M. Rossini, S. Adami, Management of patients with complex regional pain
imperfecta, Nat. Rev. Endocrinol. 7 (2011) 540–557, http://dx.doi.org/10.1038/ syndrome type I, Osteoporos. Int. J. 27 (2016) 2423–2431 Establ Result Coop Eur
nrendo.2011.81. Found Osteoporos Natl Osteoporos Found USA. (doi: 10.1007/s00198-016-3531-9).
[3] H. Hoyer-Kuhn, C. Netzer, O. Semler, Osteogenesis imperfecta: pathophysiology and [30] J.P. Devogelaer, J. Malghem, B. Maldague, C. Nagant de Deuxchaisnes, Radiological
treatment, Wien. Med. Wochenschr. 165 (2015) 278–284 1946. (doi: 10.1007/ manifestations of bisphosphonate treatment with APD in a child suffering from os-
s10354-015-0361-x). teogenesis imperfecta, Skelet. Radiol. 16 (1987) 360–363.
[4] P. Trejo, F. Rauch, Osteogenesis imperfecta in children and adolescents-new devel- [31] T. Palomo, F. Fassier, J. Ouellet, et al., Intravenous bisphosphonate therapy of young
opments in diagnosis and treatment, Osteoporos. Int. 27 (2016) 3427–3437 J Establ children with osteogenesis imperfecta: skeletal findings during follow up through-
Result Coop Eur Found Osteoporos Natl Osteoporos Found USA. (doi: 10.1007/ out the growing years, J. Bone Miner. Res. Off. J. Am. Soc. Bone Miner. Res. 30
s00198-016-3723-3). (2015) 2150–2157, http://dx.doi.org/10.1002/jbmr.2567.
L. Idolazzi et al. / Bone 103 (2017) 144–149 149

[32] K. Lindahl, A. Kindmark, C.-J. Rubin, et al., Decreased fracture rate, pharmacogenetics Bone Miner. Metab. 30 (2012) 434–438, http://dx.doi.org/10.1007/s00774-011-
and BMD response in 79 Swedish children with osteogenesis imperfecta types I, III 0331-3.
and IV treated with Pamidronate, Bone 87 (2016) 11–18, http://dx.doi.org/10.1016/j. [35] N. Nicolaou, Y. Agrawal, M. Padman, et al., Changing pattern of femoral fractures in
bone.2016.02.015. osteogenesis imperfecta with prolonged use of bisphosphonates, J. Child. Orthop. 6
[33] A.A. Hennedige, J. Jayasinghe, J. Khajeh, T.V. Macfarlane, Systematic review on the in- (2012) 21–27, http://dx.doi.org/10.1007/s11832-011-0380-0.
cidence of bisphosphonate related osteonecrosis of the jaw in children diagnosed [36] M. Rossini, G. Adami, S. Adami, et al., Safety issues and adverse reactions with oste-
with osteogenesis imperfecta, J. Oral Maxillofac. Res. 4 (2013), e1. http://dx.doi.org/ oporosis management, Expert Opin. Drug Saf. 15 (2016) 321–332, http://dx.doi.org/
10.5037/jomr.2013.4401. 10.1517/14740338.2016.1136287.
[34] E. Maines, E. Monti, F. Doro, et al., Children and adolescents treated with neridronate
for osteogenesis imperfecta show no evidence of any osteonecrosis of the jaw, J.

You might also like