Sunteți pe pagina 1din 7

International Journal of Pharmaceutics 477 (2014) 32–38

Contents lists available at ScienceDirect

International Journal of Pharmaceutics


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

Study of quality and stability of ursodeoxycholic acid formulations for


oral pediatric administration
A. Santoveña *, E. Sánchez, L. Charola, M. Llabrés, J.B. Fariña
Departamento de Ingeniería Química y Tecnología Farmacéutica, Facultad de Farmacia, Universidad de La Laguna, 38200 La Laguna, Tenerife, Spain

A R T I C L E I N F O A B S T R A C T

Article history: This paper describes a rational method of characterizing the biopharmaceutical stability of two oral
Received 8 September 2014 suspensions of ursodeoxycholic acid (UDCA) used in pediatrics. Because there is no commercial
Received in revised form 2 October 2014 presentation of UDCA that can administer appropriate doses for infants and children, an active
Accepted 4 October 2014
pharmaceutical ingredient (API) formulation is required. Due to its very low solubility and low dose in the
Available online 8 October 2014
formula (1.5%), two different suspensions with minimal use of excipients were studied, avoiding the use
of complex additives and those not recommended by the European Medicines Agency (EMA). Adherence
Keywords:
to Standard Operating Procedure (SOP) allows the preparation of formulations with appropriately sized
Ursodeoxycholic acid oral suspension
Pediatric administration
and stable particles, and suitable rheological behavior in withdrawing the dose after stirring. Dose
Standard Operating Procedure uniformity, expressed as mass and content variability, was determined using the criteria of the European
Dose uniformity and the United States Pharmacopoeia. Additionally, dose content variation of every mass determined was
Stability studied. A rational method was developed for determining the dose uniformity of UDCA in suspensions,
whether freshly prepared or after storage under different conditions for 30 and 60 days. This method
permits detection of differences between doses taken at different heights in the vessel at various times
and storage conditions. UDCA was stable under all conditions studied, requiring the presence of glycerol
in the formulation to obtain the declared API value after stirring. Storage of UDCA suspensions in a
refrigerator increased variability between doses.
ã 2014 Elsevier B.V. All rights reserved.

1. Introduction and the treatment of parenteral nutrition-associated cholestasis


(De Marco et al., 2006). The recommended UDCA dose for children
Ursodeoxycholic acid (UDCA) is a white crystalline powder is 5–30 mg/kg/day divided into two or three doses (Galabert et al.,
that is poorly soluble in water and highly permeable (Buck, 2009), 1992; Arslanoglu et al., 2008). UDCA is available depending on
so it belongs to class II drug products in the Biopharmaceutical the country as commercial capsules (150–500 mg) or tablets
Classification System (Jadhav et al., 2012). UDCA is a naturally (150–500 mg) (Vademecum, 2010). However, there are no
occurring bile acid that is physiologically produced in the liver commercial liquid formulations for patients who can not swallow
and present in small amounts in human bile. It suppresses the capsules or tablets, such as infants and children (Johnson and
synthesis and secretion of endogenous cholesterol by the liver Streetman, 2002). Pharmaceutical suspensions can however be
and inhibits intestinal absorption of cholesterol. It is a hydrophilic made in a hospital or pharmacy to administer the drug to such
bile acid that solubilizes cholesterol, promoting dispersion into patients. There are few published studies on the stability of oral
body fluids, reducing its viscosity and increasing bile flow. suspensions of UDCA (Johnson and Streetman, 2002; Mallett et al.,
Consequently, UDCA reduces cholestasis, preventing the forma- 1997; Geiger et al., 2012). All of these use the active pharmaceutical
tion and promoting dissolution of cholesterol gallstones (Buck, ingredient (API), by modifying a commercially available tablet or
2009). capsule. Currently, this technique is not allowed by legislation in
UDCA has been used in infants and children with different some countries due to the use of the marketed drug outside its
pathologies: hepatic and biliary cholestasis, as adjuvant therapy instructions or introducing excipients or amounts of them not
(Balisteri, 1997), management of biliar atresia (Ullrich et al., 1987), allowed for pediatric oral formulations. Using high performance
liquid chromatography (HPLC), Johnson and Streetman (2002)
assessed the stability of suspensions prepared from commercial
* Corresponding author. Tel.: +34 922 316502x6812; fax: +34 922 318506. tablets. This study proved that the formulation was stable when
E-mail address: ansanto@ull.es (A. Santoveña). stored for 90 days refrigerated and at room temperature without

http://dx.doi.org/10.1016/j.ijpharm.2014.10.011
0378-5173/ ã 2014 Elsevier B.V. All rights reserved.
A. Santoveña et al. / International Journal of Pharmaceutics 477 (2014) 32–38 33

change in odor, color, taste, or visible signs of microbial growth - firstly, the methylcellulose vehicle is prepared.
(Johnson and Streetman, 2002). Another assay concluded that - all the solid components are pulverized and weighed.
UDCA 25 mg/ml was stable for 60 days in an oral liquid prepared - the UDCA (1.5 g) is added to a 100 ml Erlenmeyer and then the
extemporaneously from capsules and sweetened vehicle, when it right amount of glycerol (F1 only) is added with constant
was stored refrigerated or at ambient temperature in amber plastic shaking, for 5 min, until a homogeneous paste is formed.
bottles (Mallett et al., 1997). More recently, Geiger et al. studied the - then, half the amount of methylcellulose solution is added with
stability of an UDCA suspension prepared free of sorbitol and constant shaking, for 5 min. The process is repeated with the
alcohol in a commercial suspending vehicle, showing it was stable other half of the solution.
for 66 days when protected from light and kept refrigerated (Geiger - half the amount of suspensor vehicle is added to the previous
et al., 2012). In view of these results, the formulations prepared mixture.
from the marketed drug do not have stability problems for the - the contents of the Erlenmeyer are transferred to a 100 ml
intended use. graduated cylinder.
A stable suspension allows the development of a liquid dosage - to recover the rest of the suspension, the flask is washed twice
form containing an appropriate quantity of drug in an acceptable with the other half of suspensor vehicle, completing the 100 ml
volume, but it is necessary to check two fundamental aspects: (a) volume.
the safety of the other excipients for pediatric patients according - it is then packaged in a 125 ml amber bottle with dispenser
to the European Medicines Agency (EMA, 1995), and (b) the closure.
concentration of the active substance – to ensure that the correct
dose is administered (Jadhav et al., 2012), especially if the active The suspensions were physically and chemically characterized
ingredient is administered in a low proportion (Sundell-Breden- as described below.
berg and Nyströn, 2001). This last point is very important because
any dose or stability testing is normally done at a pharmacy or 2.2. Quality control
hospital due to the effort required to perform a stability-
indicating HPLC for every formulation prepared (Glass and 2.2.1. pH
Haywood, 2006). For this reason, there are no studies either The pH was tested in duplicate using a Crison GLP 21 pHMeter
about the quality of the final product or the administered dose (Barcelona, Spain).
homogeneity. It may be necessary to apply a pharmaceutical
quality system such as ICH Q10 (2014) to ensure that the proposed 2.2.2. Particle size
Standard Operating Procedure (SOP) lets us take the correct dose The particle size was tested in duplicate by a Mastersizer1 2000
of active ingredient from the formulation by the method (Malvern, UK).
indicated by the manufacturer (syringe, spoon, cup). Owing to
the above situation, the aim of this study was the design of a 2.2.3. Rheology
pediatric oral suspension with a low proportion of pure UDCA and The storage (G0 ) and loss (G00 ) moduli of each formulation at
the development of a simple and feasible methodology that different times were measured using a Bohlin1 rheometer CVOD
ensures that the suspensions prepared contain a uniformly 100 (Malvern Instruments Ltd., Malvern, UK) equipped with Bohlin
distributed API throughout their volume. Consequently the v06.51 Software in oscillation tests. The experiments were carried
extracted doses, determined as dose mass and content out in triplicate in the linear region of the material and with a
variation, will be within the limits allowed for United States plate-plate 20 (PP 20) measuring system.
and European Pharmacopoeia for other oral dosage forms.
This need especially applies to those suspensions with a 2.2.4. HPLC method validation
ow proportion of an active ingredient with low aqueous The UDCA content in the suspension was measured by reversed
solubility. phase chromatography (RP-HPLC) following the method described
below.
2. Materials and methods The chromatographic system (Waters, Millford, MA, USA)
consisted of a pump, a Model 600E Multisolvent delivery system,
UDCA and excipients were pharmacopoeia grade, provided
by Acofarma (Spain). All other reagents were analytical grade
(Sigma–Aldrich, Spain).

2.1. General SOP

Two different formulations were used to prepare the suspen-


sions, see Table 1. At least three batches of each formulation were
made.
The suspensions were elaborated according to the following
SOP:

Table 1
Characterization of UDCA suspensions.

F Wetting Suspending agent (1% w/w) pH Particle size (mm)


agent
1 Glycerol 20% Methylcellulose 1000 4.4  0.4 99.3  0.1
2 – Methylcellulose 1000 6.0  0.1 98.7  0.1

F, formulation; mean value  SD; SD, standard deviation. Fig. 1. Control chart for proposed UDCA RP-HPLC method.
34 A. Santoveña et al. / International Journal of Pharmaceutics 477 (2014) 32–38

Fig. 2. UDCA pure pattem chromatographic peak (continuous line) and at 2 days at
pH 8 after the drug was stored at 33  C and 70  C. The plot of the results of the assay
at 70  C is up-shifted 0.005 units on the Y-axis.

a 717 plus Autosampler, a 2414 refractive index (RI), and a NovaPack


C18 column (3.9 mm  150 mm) packed with 4 mm particles as
stationary phase. The data acquisition software used was Millenium
32 (Chromatographic Manager, Waters Corporation). The mobile
phase was a mixture of 0.1% (v/v) acetic acid/methanol 30/70 (v/v) at
a flow rate of 0.8 ml/min at 40  C (Peepliwal et al., 2011). All chemicals
and reagents were HPLC grade. All solvents were filtered with
0.45 mm pore-size filters (Millipore, Billerica, MA, USA). The mobile
phase was filtered and degassed.
In order to validate the analytical method (ICH Q2, 2014), seven
UDCA standard solutions were prepared at concentrations of
20–80 mg/ml. Every sample was analyzed five times. The variance
analysis (ANOVA) of the linear regression confirmed the linearity
of the method, through rejection of the null hypothesis of linearity
deviation for a significance level of 0.05 (a = 0.05); the coefficient of
variation of the method was 3.8%. The equation of the regression
line was: area = 8181 + 4297  C; r = 0.999 (n = 35), with a residual
standard error of 8876. The method precision (as repeatability) was
0.93%, it was determined by a six-fold analysis of the same sample.
System accuracy was expressed as percentage recovery by the
assay of a known added amount of drug, the mean value being
102% (n = 9). The detection and quantitation limits, based on the
Fig. 3. Variation in the shear stress with the shear rate a), and the complex viscosity
standard deviation of the response and slope, were 6.5 and
with the shear rate b) of F1 and F2.
19.7 mg/ml respectively.
A robustness test was performed to examine the effect of
operational parameters on the analysis results. The flow-rate 2.2.5. Extraction yield
(0.8  0.01 ml/min), injection volume (25  1 ml), temperature To determine the efficiency of extraction, twenty samples of
(40.0  1.5  C), mobile phase composition (30  1 for acetic acid, 5 ml each were taken, to which had been added 75 mg of UDCA and
70  1 for methanol, v/v), and column performance over time were the quantities of each of the formulation components. After
determined in order to confirm the method’s robustness. To homogenization of the samples, they were dissolved in 25 ml of
calibrate the RP-HPLC system and monitor its performance, we methanol to ensure complete dissolution of UDCA. Subsequently,
analyzed an UDCA solution sample daily as standard. The they were suitably diluted with the mobile phase to proceed with
estimated area for standard concentration was 223,031 with an chromatographic analysis.
RSD of 2.4%. The upper and lower limits for the control chart
were established at 3SD of this value, taking as SD the value 2.2.6. Dose content uniformity
obtained from the variance of the analytical method. Fig. 1 shows For USP, the compounded preparations have to be prepared,
the control chart for the method, where the peak’s area stays ensuring that every preparation contains no less than 90% and no
between the established limits every time. The chromatographic more than 110% of the theoretically calculated and labeled amount
conditions (e.g., flow-rate, relative mobile phase composition) of active ingredient per unit weight or volume (USP, 2007). The
and column performance were checked, especially the tailing European Pharmacopoeia (EP) indicates that for multi-dose
factor and column efficiency. When necessary, corrective action containers, 20 doses are individually weighed at random and
was taken. the individual and average masses are determined. No more than
A. Santoveña et al. / International Journal of Pharmaceutics 477 (2014) 32–38 35

until the concentration interval of the standard solutions was


studied. This dose volume was taken out with an oral syringe after
the suspension was shaken (Wening and Breitkreutz, 2011). An
UDCA mass balance was determined from the volume remaining in
the containers after removal of 20 doses.
A multicenter study was performed with the proposed SOP for F2,
wherein the pharmacy services of three public hospitals (H1, H2 and
H3) collaborated, along with a compounding pharmacy (CP).
An additional test was performed to determine the uniformity
of the dose extracted from different heights of the elaborated
suspensions. Each formulation was poured into a suitable
container to study the degree of flocculation and sedimentation
of the suspensions (100 ml graduated cylinder) and after stirring
(10 times inverted 180 ), doses were taken at upper, middle and
lower heights. After a certain time at rest (1.5 min), the variation in
these values taken at the same heights was found. Doses taken
were weighed and their UDCA contents determined. This process
was repeated at 15, 30 and 60 days in different storage conditions
described below.

2.2.7. Stability study


The formulations were checked for stability, at different
temperatures and storage conditions. At 5  C (Fridge-stove
P-selecta Welidow type, Spain), at 23  C (dark chamber, own
manufacture), and at 40  C (Heraeus UT 6060, Spain). One
photostability study was also performed at 25  C in a UV–vis
chamber that meets the specifications of the ICH (Q1B), consisting
of a cool white lamp that meets the criteria ISO 10977 (1993), along
with a UV lamp emitting between 320 and 400 nm. The trials lasted
60 days, except for the F1 tests at 5  C and 23  C, which ended at
30 days. Two extreme stability tests were also conducted at 33 and
70 C at pH 8.0. Three samples of 5 ml from every UDCA
formulation batch were taken at predetermined intervals from
different heights (as described above).

3. Results and discussion

The use of marketed UDCA was ruled out due to the legal and
safety limitations described above. A class II drug product
compounded at a low dose (1.5% w/v) in suspension requires
different agents in quantities that ensure adequate contact
Fig. 4. Variation in complex viscosity after different conditions of F1 a) and F2 b). between the solid and the rest of the liquid preparation. This
guarantees correct drug suspension and administration.
2 of the individual masses deviate from the average by more than The most commonly employed dose for UDCA is 1.5% (w/v)
10% and none deviates 20% (RFE, 2005). (Atienza et al., 2011). We propose including an excipient (such as
Twenty doses (5 ml) taken randomly from two elaborated glycerol) in the formulation to facilitate wetting of the solid in the
formulations were weighed prior to determining each UDCA viscous medium in which a class II API is dispersed.
content. Afterwards, the samples were diluted with mobile phase Fig. 2 shows the chromatogram of pure UDCA obtained by the
RP-HPLC method. Only one peak with an elution volume of 6.6 ml
Table 2 was detected. In the same figure we can see the shape of the
Evolution of UDCA% in time for F1 and F2 formulations at different storage chromatogram after two days storage at 33  C and 70  C at pH 8.
conditions. Only in this extreme environment, an additional chromatographic
F1 UDCA % species appears at 4.5 ml and the peak appears with tail at 33 and
70  C respectively.
t (days) 5 C 23  C 25  C 40  C
The average extraction yield of UDCA from the suspension by
0 92.7  9.8 92.7  9.8 92.7  9.8 92.7  9.8 RP-HPLC method was 95.1  0.06%.
15 102  29 95.7  1.1 – 90.0  2.9
30 101  19 92.9  2.8 90.2  8.9 92.1  3.3
Table 1 shows the composition of the different suspensions,
60 ND ND 101  8.7 99  4.2 such as the initial pH and particle size. As discussed above, the low
UDCA proportion (1.5% w/v) justifies the use of a wetting agent like
F2 UDCA % glycerol in F1 to assess its influence on final quality. Both
t (days) 5 C 23  C 25  C 40  C formulations showed the same particle size, near 100 mm. When
0 98.4  3.0 98.4  3.0 98.4  3.0 98.4  3.0 stored at 40  C for 60 days (F1) and 30 days (F2), the size calculated
15 92.5  52 97.3  34 105  42 97.1  1.5 was 99.9 and 82.3 mm respectively. Only F2 showed a slight
30 101  3.7 95.9  9.4 94.7  6.0 99.9  3.2 decrease in particle size in these conditions.
60 100  12.6 97.8  4.8 98.0  2.0 96.6  4.4 Both formulations showed non-Newtonian behavior wherein
ND, not detected. the viscosity of each system decreases with an increasing shear-
36 A. Santoveña et al. / International Journal of Pharmaceutics 477 (2014) 32–38

Table 3
Mass uniformity test in all formulations studied.

Dw (mg)

F1 F2 F2-H1 F2-H2 F2-H3 F2-CP

1 5.62 5.82 5.73 5.73 5.79 5.72


2 5.58 5.85 5.80 5.69 5.77 5.75
3 5.68 5.88 5.83 5.78 5.78 5.77
4 5.71 5.86 5.86 5.77 5.77 5.78
5 5.75 5.82 5.87 5.73 5.79 5.77
6 5.76 5.82 5.81 5.74 5.79 5.72
7 5.70 5.85 5.85 5.79 5.81 5.73
8 5.67 5.86 5.80 5.80 5.79 5.72
9 5.67 5.86 5.81 5.76 5.79 5.74
10 5.67 5.86 5.81 5.80 5.78 5.72
11 5.69 5.92 5.86 5.04 5.80 5.78
12 5.61 5.84 5.80 4.90 5.82 5.78
13 5.71 5.86 5.85 5.65 5.80 5.77
14 5.68 5.84 5.87 5.73 5.85 5.78
15 5.65 5.86 5.85 5.76 5.85 5.78
16 5.66 5.85 5.81 5.76 5.85 5.75
17 5.67 5.84 5.86 5.71 5.85 5.74
18 5.69 5.83 5.84 5.74 5.84 5.74
19 5.36 5.86 5.83 5.37 5.85 5.77
20 5.67 5.85 5.85 5.78 5.85 5.79
A 5.66 5.85 5.83 5.65 5.81 5.75
LL UL LL UL LL UL LL UL LL UL LL HL
10% 5.09 6.23 5.27 6.44 5.25 6.41 5.09 6.22 5.23 6.39 5.23 6.39
20% 4.53 6.79 4.68 7.02 4.66 7.00 4.52 6.78 4.65 6.97 4.65 6.97

Dw, dose weight; A, average; LL, lower limit; UL, upper limit; H, hospital; CP, compounding pharmacy; Shading values, outside the limits of 10%.

rate (Fig. 3). The non-Newtonian suspensions were fitted to 5  C for 40 days and at 40  C for 30 days show a dramatic decrease
different mathematical models and they behaved as Bingham in viscosity, from 73.3 to 2.53 Pa s and 1.46 Pa s, respectively. The
materials, as do other oral suspensions (Gao et al., 2004; Santoveña viscosity of both increased after a settling period following stirring.
et al., 2010), with a yield value of about 1.0 Pa s. This slows the sedimentation rate of suspended particles and
Fig. 4a shows the variation in viscosity of complex F1 at delays the formation of sediment. After stirring, the viscosity
frequency 1 Hz at time zero, after stirring, 45 min of settling, decreases and facilitates homogeneous removal of the UDCA dose.
and 60 days of storage at 40  C. The viscosity remained constant Unlike F2, F1 maintained its viscosity for 60 days at 40  C. The
after storage at 40  C, taking values at 1 Hz of 18.6 and 15.6 Pa s, difference between the two formulations is the incorporation of
respectively. The viscosity increased to 63.6 Pa s after 45 min at glycerol (F1), which reduces the contact angle between the particle
rest. Like F1, the F2 viscosity (Fig. 4b) increases after 45 min at rest and the suspending medium. This keeps the particles in suspen-
(from 73.3 to 227 Pa s). But in this case, the formulations stored at sion longer and maintains the viscosity of the formulation during

Table 4
Content uniformity test in all formulations studied.

DV%

F1 F2 F2-H1 F2-H2 F2-H3 F2-CP

1 84.7 103 39.5 37.2 13.2 37.6


2 73.4 115 41.8 22.8 32.0 33.2
3 77.4 110 42.4 24.7 14.4 35.9
4 87.6 117 47.9 29.6 30.0 51.9
5 92.8 109 45.8 68.7 21.5 44.2
6 94.0 115 52.3 35.5 24.7 56.2
7 90.1 115 64.3 140 82.4 75.2
8 95.3 109 73.2 78.8 89.1 60.3
9 97.1 100 88.5 86.3 84.2 70.0
10 89.3 97.7 85.8 101 97.9 49.2
11 92.6 179 33.2 41.8 127 50.3
12 94.8 110 21.3 51.4 129 52.2
13 100 110 22.4 65.1 145 58.2
14 95.4 98.5 25.7 64.2 33.3 60.7
15 94.5 99.9 23.1 53.6 20.5 69.5
16 95.6 96.8 33.6 81.9 43.4 41.4
17 89.5 92.8 46.5 112 49.2 39.6
18 98.1 91.5 47.9 91.2 56.4 40.9
19 95.9 95.1 53.4 90.3 76.4 43.3
20 89.0 94.9 53.0 105 89.0 46.7
A 91.4 108 47.1 69.1 62.9 50.8
LL UL LL UL LL UL LL UL LL UL LL UL
10% 82.2 100.5 97.1 118.7 42.4 51.8 62.2 76.0 56.6 69.2 45.7 55.9
20% 73.1 109.7 86.4 129.5 37.7 56.5 55.2 82.9 50.3 75.5 40.6 61.0

Dw, dose weight; A, average; LL, lower limit; UL, upper limit; H, hospital; CP, compounding pharmacy; Soft shading values, outside the limits of 10%; Strong shading, outside
the limits of 20%.
A. Santoveña et al. / International Journal of Pharmaceutics 477 (2014) 32–38 37

Table 5 To confirm the effect of glycerol on the API distribution and to


Results of two-factor ANOVA with one sample per group for F1 and F2.
assay variation in uniformity of doses taken from different heights
F1 Probability of the same formulation, 2-factor ANOVA (time and storage
Time Storage conditions
conditions) was performed. For this, two parameters were
studied: Dmax, the maximum difference between doses taken
Dmax
DV% 0.9816 0.1248
at different heights and Dt, the Dmax between two sampling
D (mg/g) 0.9496 0.1034 times. Dmax and Dt were expressed as percentage of declared
dose (DV%) and amount of UDCA by dose weight (D mg/g),
Dt respectively.
DV% 0.0006 0.2831
As can be seen in Table 5, for both formulations there were no
D (mg/g) 0.0024 0.2090
statistically significant differences (p > 0.05) between storage
F2 Probability conditions for any of the factors studied. However, there were
Time Storage conditions
significant differences in the extracted dose between times, except
for Dmax in F1. Thus, F1 is the most stable and highest quality
Dmax
DV% 0.0134 0.3118
formulation because no statistically significant differences were
D (mg/g) 0.0162 0.3177 detected between the doses taken at different heights (Dmax), at
different times and storage conditions. But for F1, the differences in
Dt Dt variation between times are statistically significant. This
DV% 0.0109 0.3142
indicates that the dose should be immediately removed after
D (mg/g) 0.0284 0.2966
agitation of the suspension, independently of the height from
Dmax, difference between the maximum and minimum dose; %DV, percentage of
which it is taken.
declared value; D (mg/g), amount of active ingredient by dosage weight; Dt,
difference between the maximum and minimum dose between two sampling
Fig. 5 shows the variability among the contents determined as
times. DV% for doses extracted at different heights and in different storage
conditions. It shows how for both F1 and F2 the greatest dose
variability is found in the formulatons stored at 5  C. Therefore,
the storage time. Table 2 shows the results of stability tests. F1 and contrary to expectations, storage in a refrigerator does not favor
F2 kept their initial UDCA concentrations between 90 and 105% for homogeneous distribution of UDCA in F1 or F2. Due to the increase
all storage conditions. In no case was there a higher variation than in viscosity of the formulation at low temperatures, homogeneity
10% of the initial concentration of UDCA. of the suspension was impaired before dose removal. Although, in
Tables 3 and 4 respectively show the mass and content either case, the doses taken always contained between 90 and 110%
uniformity tests performed for F1 and F2. Both formulations of the initial dose tested.
met the EP test for uniformity of mass, that is, no more than 2 of the
individual masses deviate from the average by more than 10% and 4. Conclusions
none deviates 20%. If we apply the same EP criteria for the declared
UDCA content determined in each preweighed dose, only F1 met The chromatographic method allowed us to detect and quantify
the test, see Table 4. Applying the USP criteria for compounding UDCA accurately and precisely, both as pure pattern and after
preparations, the average declared content for both F1 and extraction from the formulations studied (F1 and F2). The
F2 prepared in our laboratory would meet the test. Those prepared suspensions showed a similar initial particle size. F1 and
in hospitals and compounding pharmacies do not meet either of F2 showed plasticity in their behavior, indicating an increase in
the Pharmacopoeias’ criteria. The dose weight of these formula- viscosity after storage at rest. F1 kept its viscosity value after being
tions was similar to those obtained with the other formulations. stored 60 days at 40  C.
Total mass balances are close to or exceed the total amount needed The suspensions prepared in our pharmaceutical technology
to prepare the suspension (data not shown). This indicates a poor laboratory had a concentration near the theoretically calculated
distribution of the active substance in the suspension due to and labeled amount of active ingredient per unit volume
improper SOP monitoring and not to a possible loss of it due to the (1.5 mg/ml), with a recovery percentage between 90 and 110%,
low dose rate used in its preparation (1.5%). even when stored at 40  C for 30 (F2) and 60 days (F1). Using
HPLC, UDCA stability was thus confirmed in suspensions stored in
the presence of light (or not) at 5, 23, 25 and 40  C for 60 days for
the usual formulation and 30 days for the other with glycerol.
Both formulations met the EP test for uniformity of mass, and
USP criteria for the average declared value of compounding
formulations. Only F1, the suspension incorporating glycerol in its
formula, complied with the same EP criteria if applied to the
declared content determined for every preweighed dose used in
the mass uniformity test.
The use of certain parameters (Dmax and Dt) in extracted dose
determination enabled us to make a statistical study of doses taken
from different heights of the formulations stored under different
conditions, and their evolution over time. This study confirms that
if the F1 suspension is made strictly following the SOP and after
stirring, UDCA doses are extracted between 90 and 110% of the
declared value from different heights and after 30 days of storage
in different conditions. Analysis of the variance in dose content
sampled at different heights of the suspension shows that
Fig. 5. Variability between F1 and F2 content determined as DV% for doses taken at refrigerator storage increases variability, unlike other temperature
different heights and different storage conditions. and storage conditions.
38 A. Santoveña et al. / International Journal of Pharmaceutics 477 (2014) 32–38

Strict compliance with the SOP in both clinical and community Gao, S., Moros, D., Moldenhauer, M.G., 2004. Patent No. (20040258716).
compounding is essential to ensure the quality, safety and efficacy Washington, DC: US Patent & Trademark Office.
Geiger, C.M., Voudrie, M.A.I.I., Sorenson, B., 2012. Stability of ursodiol in SyrSpend SF
of the formulation prepared. Awareness of this is essential in daily cherry flavored. Pharm. Compd. 6, 510–512.
practice, especially when it comes to infant and child patients. Glass, B.D., Haywood, A., 2006. Stability considerations in liquid dosage forms
extemporaneously prepared from commercially available products. J. Pharm.
Pharm. Sci. 9, 398–426 http://www.ich.org/fileadmin/Public_Web_Site/ICH_-
Acknowledgements Products/Guidelines/Quality/Q10/Step4/Q10_Guideline.pdf (accessed 04.07.14).
ICH Q10, 2014. The International Conference on Harmonisation.
The authors are very grateful for their collaboration to the ICH Q2(R1), 2014. The International Conference on Harmonisation. http://private.
ich.org/LOB/media/MEDIA417.pdf (accessed 20.06.14).
hospital pharmacy services of Hospital Universitario Nuestra Jadhav, K.R., Sankpal, S.V., Sawant, S.V., Kadam, V.J., 2012. Development and in vitro
Señora de la Candelaria, Hospital Universitario de Canarias and evaluation of film coated micronized inmediate release tablets of ursodeox-
Complejo Hospitalario Insular-Materno Infantil, Farmacia Feria, ycholic acid. Der Pharmacia Lettre 4, 714–719.
Johnson, C.E., Streetman, D.D., 2002. Stability of oral suspensions of ursodiol made
Instituto Tecnológico del Medicamento Individualizado (ITMI) and
form tablets. Am. J. Health Syst. Pharm. 59, 361–363.
Comisión Nacional de Unificación de Criterios en Formulación Mallett, M.S., Hagan, R.L., Peters, D.A., 1997. Stability of ursodiol 25 mg/ml in an
Pediátrica. extemporaneously prepared oral liquid. Am. J. Health Syst. Pharm. 54,
1401–1404.
Peepliwal, A., Bonde, C.G., Bothara, K.G., 2011. A validated RP-HPLC method for
References quantitative determination of related impurities of ursodeoxycholic acid (API)
by refractive index detection. J. Pharm. Biomed. Anal. 25, 845–849.
Arslanoglu, S., Moro, G.E., Tauschel, H., Boehm, G., 2008. Ursodeoxycholic acid RFE, uniformidad de masa de las preparaciones presentadas en multidosis (01/
treatment in preterm infants: a pilot study for the prevention of cholestasis 2005, 20901), 2005. Ministerio de Sanidad y Consumo, Agencia Española de
associated with total parenteral nutrition. J. Pediatr. Gastroenterol. Nutr. 46, Medicamentos y Productos Sanitarios, eds. Real Farmacopea Española 3a
228–231. edición, Madrid 289.
Atienza, M., Martínez, J., Álvarez, C., 2011. Formulación en farmacia pediátrica. Santoveña, A., Llabrés, M., Fariña, J.B., 2010. Quality control and physical and
Madrid Vicente. chemical stability of hydrocortisone oral suspension: an interlaboratory study.
Balisteri, W.F., 1997. Bile acid therapy in pediatric hepatobiliary disease: the role of Int. J. Pharm. Compd. 14, 430–435.
ursodeoxycholic acid. J. Pediatr. Gastroenterol. Nutr. 24, 573–589. Sundell-Bredenberg, S., Nyströn, C., 2001. The possibility of achieving an interactive
Buck, M.L., 2009. Use of Ursodiol in infants and children. Pediatr. Pharmacother. 15, mixture with high dose homogeneity containing an extremely low proportion
1–4. of a micronized drug. Eur. J. Pharm. Sci. 12, 285–295.
De Marco, G., Sordino, D., Bruzzese, E., Di Caro, S., Mambretti, D., Tramontano, A., The United States Pharmacopeial Convention, 2007. Compounded Preparations. The
Colombo, C., Simoni, P., Guarino, A., 2006. Early treatment with ursodeoxycholic United States Pharmacopeia 30-The Nacional Formulary 25, vol. 1 (795) 2007.
acid for cholestasisin children on parenteral nutrition because of primary Ullrich, D., Rating, D., Schroter, W., Hanefeld, F., Bircher, J., 1987. Treatment with
intestinal failure. Aliment. Pharmacol. Ther. 24, 387–394. ursodeoxycholic acid renders children with biliary atresia suitable for liver
European Medicines Agency Science Medicine Health, 1995. EMA. http://www.ema. transplantation. Lancet 2, 1324.
europa.eu/docs/en_GB/document_library/Scientific_guideline/2009/09/ Ursodesoxicólico ácido, 2010. Vademecum, Vidal Vademecum Spain. http://www.
WC500003412.pdf (accessed 20.06.14). vademecum.es/principios-activos-ursodeoxicolico+acido-a05aa02 (accessed
Galabert, C., Montet, J.C., Leugrand, D., Lecuire, A., Sotta, C., Figarella, C., Chazalette, J. 20.06.14).
P., 1992. Effects of ursodeoxycholic acid treatment on liver in patients with Wening, K., Breitkreutz, J., 2011. Oral drug delivery in personalized medicine: unmet
cystic fibrosis and chronic cholestasis. J. Pediatr. 121, 138–141. needs and novel approaches. Int. J. Pharm. 404, 1–9.

S-ar putea să vă placă și