Buchan 2012

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 10

RESEARCH ARTICLE

Suppository Formulations as a Potential Treatment


for Nephropathic Cystinosis
BARBARA BUCHAN, GRAEME KAY, KERR H. MATTHEWS, DONALD CAIRNS
Institute for Health & Welfare Research, Robert Gordon University, Aberdeen, UK

Received 22 February 2012; revised 2 May 2012; accepted 8 June 2012


Published online in Wiley Online Library (wileyonlinelibrary.com). DOI 10.1002/jps.23246

ABSTRACT: Nephropathic cystinosis is a rare autosomal recessive disease characterised


by raised lysosomal levels of cystine in the cells of all the organs. It is treated by the 6-h
oral administration of the aminothiol, cysteamine, which has an offensive taste and smell.
In an attempt to reduce this frequency and improve the treatment, cysteamine-containing
polyethylene glycol suppositories were prepared and evaluated for dissolution and stability. The
results demonstrated that cysteamine release was complete after 30 min, and that there was a
uniform drug distribution within the formulations. Twelve-month stability tests highlighted a
potential incompatibility among some excipients, although stability was demonstrated for the
cysteamine suppositories up to 6 months. These suppositories may provide a useful alternative
to the current oral therapy for cystinosis. © 2012 Wiley Periodicals, Inc. and the American
Pharmacists Association J Pharm Sci
Keywords: Cystinosis; Formulation; PEG; Physical Stability; Dissolution; Thermal Analysis;
Pediatric

INTRODUCTION renal failure by age nine,5 and grow at 50%–60% of


the expected rate. By the age of eight, an untreated
Nephropathic cystinosis is a rare genetic disease char-
child with cystinosis will be of the height of a 4-year
acterised primarily by extremely high intracellular
old,5 and, if inadequately treated, may not reach 4 ft.
levels of the amino-acid cystine, electrolyte imbal-
in height.
ance, proximal renal tubular dysfunction (Fanconi
The condition is caused by a defect in the CTNS
syndrome) and general failure to thrive.1,2 The accu-
gene. This is a gene which codes for a 367-amino-
mulation of cystine as crystals in most tissues leads
acid-lysosomal-transport protein called Cystinosin.
to the progressive dysfunction of multiple organs.3
In healthy cells, the amino-acid cystine is transported
The incidence of cystinosis is one in 100,000–200,000
into the cytoplasm of the cell for processing. In cysti-
live births, and affects approximately 2000 patients in
nosis, however, it accumulates within the lysosomes
the world, although there are believed to be many
to levels of 10–1000 times those seen in healthy
more cases undiagnosed.2,4 Some infants will die be-
cells, whereupon it crystallises from solution, caus-
cause of dehydration and electrolyte imbalance from
ing cell death and organ failure.6 This crystallisa-
Fanconi syndrome without diagnosis.5 In the US
tion is widespread throughout the majority of the
alone, there are 500–600 reported cases, with between
tissues and organs in the body.7 If left untreated,
20 and 40 born each year.5
symptomatic deterioration leads to death by the sec-
Cystinosis is categorised as a lysosomal storage dis-
ond decade of life.8
order (LSD), which is a group of progressive disorders
The treatment for cystinosis involves the adminis-
that share multi-organ failure as an endpoint.3,5 As
tration of the aminothiol cysteamine, which is used
with all LSDs, no signs of abnormality are displayed
therapeutically in the bitartrate form as CystagonTM
at birth; the first symptoms of glomerular dysfunc-
(Mylan Laboratories, USA). With early and diligent
tion begin to appear at around 6–12 months of age.2
therapy, cystinosis patients can prevent or delay
Without treatment most children will reach end-stage
most of the non-renal complications and extend their
lives into a fourth or fifth decade, live independently
Correspondence to: Graeme Kay (Telephone: +44-1224-262548;
Fax: +44-1224-262555; E-mail: g.kay@rgu.ac.uk)
and some women have borne children.5,9 The re-
Journal of Pharmaceutical Sciences
nal damage is continuous; however, the decline is
© 2012 Wiley Periodicals, Inc. and the American Pharmacists Association inevitable.5

JOURNAL OF PHARMACEUTICAL SCIENCES 1


2 BUCHAN ET AL.

Cysteamine causes a range of side effects and this By avoiding the first-pass metabolism of cysteamine,
is largely because of the high dose which is required a lower dose should be achievable, whereas the taste
as much of the drug is lost to first-pass metabolism.9 and upper-gastric side effects should be eliminated.
High blood levels must be achieved, as a large pro- A study conducted by van’t Hoff et al. was previously
portion of the administered drug will bind to cir- undertaken where a cysteamine-loaded suppository
culating proteins, and cannot be up taken by the gel, for use in cystinosis, was evaluated. However, this
cells.10,11 Patients should aim to take their dose at rectal formulation was eliminated before cysteamine
regular 6-h intervals for the treatment to work effec- absorption was completed.17
tively, as the plasma half-life of cysteamine is 1.88 h,7
with blood levels peaking at 1 h, and rapidly declin-
MATERIALS AND METHODS
ing thereafter.5 This is a lifelong commitment and
requires the patient to wake in the middle of the Materials
night.9 The drug has a foul taste and smell akin to
Cysteamine hydrochloride and polyethylene glycol
rotten eggs, which regularly induces vomiting after
(PEG) grades 400, 600, 1000, 1500, 3000, 4000,
ingestion.9 In approximately 10%–15% of patients,
6000, 8000 and 14,000 were obtained from Sigma
this can be severe enough to halt therapy.5 Cys-
(St. Louis, Missouri). Witepsol W35 was obtained
teamine and its metabolites are excreted in breath
from Gattefosse (St-Priest, France). Gelucire 39/01
and sweat, and this is also an issue, especially when
was purchased from Sasol GmbH (Witten, Germany).
the child enters education. Cysteamine has the poten-
Poloxamer F68 was bought from BASF SE (Lud-
tial to cause potentially serious stomach irritations
wigshafen, Germany). Ellman’s Reagent, 5,5 -dithi-
such as gastric acid hypersecretion, reverse peristal-
obis(2-nitrobenzoate) (DTNB) was purchased from
sis and bile reflux, and 97% of the patients report gas-
Molekula (Gillingham, UK). Tris buffer and Tween
trointestinal symptoms,12,13 which in many patients
80 were bought from Fisher (Loughborough, UK).
is severe enough to significantly limit the therapy.14–16
Compliance can therefore be a major barrier to effec- Synthesis of N,N-(Bis-L-Phenylalanyl)Cystamine
tive treatment, and lead to significant morbidity. Bistrifluoroacetate (Phenylalanine Conjugate)
The rectal route of administration can largely avoid
Cysteamine does not possess a chromophore and
the phenomenon of first-pass effect. This results from
therefore is ultraviolet (UV) transparent; thus, mon-
one of the three rectal veins (upper vein) draining into
itoring its release from formulations is very difficult.
the hepatic system, whereas the middle and lower
Initially, a phenylalanine conjugate was developed to
veins bypass this and drain directly into the systemic
tag the molecule, allowing quantitative determina-
circulation. If the suppository is positioned correctly,
tion of release of the active from the dosage form
the drug should not be subjected to the first-pass ef-
via UV spectroscopy (Fig. 1). Cystamine, the oxidised
fect. This potentially allows a smaller dose to be ad-
disulphide of cysteamine, was used in the synthe-
ministered, thereby reducing or eliminating some of
sis. The phenylalanine conjugate was subsequently
the unpleasant side effects. They may also be benefi-
replaced with cysteamine hydrochloride with DTNB
cial for treating conditions in infancy, when capsules
detection (see the section Dissolution Studies).
are difficult to administer, or when the oral route is
To a stirring solution of cystamine dihydrochlo-
compromised. Rectal formulations are useful tools,
ride (1 g, 0.00444 mol) in anhydrous dichloromethane
particularly in a case such as this where the taste
(DCM) (20 cm3 ) at room temperature, 1,8-diazabi-
and side effects render the task of swallowing a tablet
cycloundec-7-ene (1.33 mL, 0.0089 mol) was added.
very unpleasant and foreboding.
The reaction mixture was then stirred at room
Aims
Formulation science may provide a way to improve the
current medication, significantly improving the lives
of sufferers and those who care for them. By eliminat-
ing the taste and frequency of administration through
alternative dosage forms, ease of administration of
cysteamine could be improved. The aim of this work
was to develop alternative formulations of cysteamine
which could reduce or eliminate some of the side
effects experienced using the current oral capsule,
thereby improving the quality of life for those affected.
An improvement in the ease of administration of cys-
teamine to infants and young children was a central Figure 1. N,N-(bis-L-phenylalanyl)cystamine bistrifluo-
objective, therefore suppositories were investigated. roacetate, (Phenylalanine conjugate).

JOURNAL OF PHARMACEUTICAL SCIENCES DOI 10.1002/jps


SUPPOSITORY FORMULATIONS AS A POTENTIAL TREATMENT FOR NEPHROPATHIC CYSTINOSIS 3

temperature for 1 h. To this, butoxycarbonyl-L- Suppository hardness was tested on an Erweka


phenylalanine-N-hydroxysuccinimide ester (3.22 g, TBH28 hardness tester (Heusenstamm, Germany).
0.0089 mol) was added. After thin layer chromato- A hardness value of at least 1.8–2 kg is considered
graphic analysis, the reaction mixture was then parti- acceptable for unmedicated suppositories.18 If the in-
tioned between DCM (20 mL) and water ( 50mL). The ert suppository blend produced unsatisfactory results
DCM extracts were washed with water (3 × 50 mL) beyond this range, no further testing was performed.
and dried over magnesium sulphate. The resultant The suppositories with the best characteristics, based
solution was applied to a silica gel chromatography on hardness tests and appearance were selected for
column (4 × 30mL) prepared with DCM. The col- further testing.
umn was initially eluted with 100% DCM followed
by 2% graduations of methanol, with the protected
Dissolution Studies
compound eluting at 9:1 DCM–methanol.
The protected compound was dissolved in triflu- Dissolution tests were performed in a six-chambered
oroacetic acid (5 mL) at room temperature. After Sotax CH-4123 dissolution bath (Basel, Switzerland).
3.5 h, the trifluoroacetic acid was removed by co- The dissolution test method followed the standard BP
evaporation with ethanol (3 × 20 mL). Trituration test for suppositories: one suppository in a stainless-
with diethyl ether followed by filtration and drying steel basket, rotating at 100 rpm in a 1 L beaker con-
(vacuum oven at 50◦ C) gave the target compound as taining medium at 37◦ C, sampling at 5 min inter-
an off-white solid. Yield: 42%. vals for 1 h (BP, 2005 ed.). The dissolution medium
Mass spectroscopy data: m/z 447.2 (100%); 224.4 was altered depending on the active used. The re-
(50%). M: C22 H32 N4 O2 S2 + ; Exact Mass: 448.2. lease was measured by UV spectroscopy. The samples
Nuclear magnetic resonance data: All analyses were returned to the bath after analysis. Supposito-
were undertaken using a Bruker Topspin Ultrashield ries containing the phenylalanine conjugate were ini-
400MHz (Billerica, Massachusetts). 1 H NMR spec- tially tested in distilled water, using UV absorbance
trum (CD3 OD) (400 MHz) *: 2.55 (m, CH2 S2 , 2H); at 256 nm corresponding to the λmax .
2.95 (m, CH2 Ar, 1H); 3.05 (m, CH2 Ar, 1H); 3.25 Cysteamine hydrochloride release can be mea-
(NH CHH, 1H); 3.45 (m, NH CHH, 1H); 3.95 (t, sured via free sulphydryl concentration through
"-CH, 1H); 7.25 (m, Ar H’s, 5H). 13 C NMR spec- the use of DTNB [Ellman’s Reagent, 5-(3-carboxy-
trum (CD3 OD) (400 MHz) *: 37.7(CH2 S2 ); 38.8 ($- 4-nitrophenyl)disulphanyl-2-nitrobenzoic acid]. Basi-
CH2 ); 39.5 (NH CH2 ); 55.8 ("-CH); 128.9 (Ar CH); cally, 1 mol of DTNB reacts with 1 mol of cysteamine
130.1 (Ar CH); 130.5 (Ar CH); 135.6 (Ar CH); 169.7 to release 1 mol of 2-nitro-5-thiobenzoate (NTB). The
(CO). Melting range: 100–104◦ C. concentration of NTB and hence the concentration of
cysteamine can be determined using UV–visible spec-
troscopy. Calibration measurements, using an analyt-
Suppository Preformulation and Manufacture
ical wavelength of 440 nm, gave a linear response,
To allow for wastage, the weight of eight supposito- R2 = 0.9991. The dissolution medium was 90%
ries was calculated for a six-form mould. The cali- deionised water and 10% 1 M Tris buffer solution at
bration value was calculated for each mould, and the pH8. Because of the susceptibility of DTNB to pho-
displacement value was calculated for each drug. The tolysis when in solution, the tank was surrounded by
bases were weighed accurately on a Mettler AE50 aluminium foil and protected from light.
analytical balance, along with the active. The bases The Higuchi equation was used to determine the
were heated and blended together, before addition of rate order of the drug release. A result below 0.45 is
either cysteamine or phenylalanine conjugate. This indicative of Fickian diffusion.19
mix was then thoroughly stirred and the resultant
liquid poured into a mould and allowed to cool. The
Active Dispersion Studies
tops were then trimmed with a spatula, and the fi-
nal forms were removed from the mould, bottled and In a suppository, there is a possibility that the tip
labelled [British Pharmacopeia (BP), 2005]. The sur- might contain more drug than the rest of the form
factant Tween 80 was used in some formulations to because of the gravitational settling.18 To ensure uni-
ensure that the fatty bases were sampled accurately form drug loading, three separate areas of the suppos-
from the dissolution medium. itory were sampled and compared using a DSC Q100
Lipophilic bases, hydrophilic bases and blends of differential scanning calorimeter (TA Instruments,
different PEG grades were examined for optimum New Castle, Delaware) and the experiment carried
characteristics. As 10 mg kg−1 was used for previous out in triplicate. Coupled with this, differential scan-
rectal delivery of cysteamine, 60 mg was used as a ning calorimetry (DSC) studies of suppositories were
nominal value.17 Until further tests are carried out carried out to determine the thermal characteristics
in vivo, the correct dose cannot be determined. of each suppository blend.

DOI 10.1002/jps JOURNAL OF PHARMACEUTICAL SCIENCES


4 BUCHAN ET AL.

Table 1. A Summary of the Batches Made and Their Characteristics

Suppository Hardness Without Hardness Including Active


Batch Composition (% w/w) Active (Cysteamine HCl) Appearance
A PEG 8000 40% 19 N, 1.94 kg 9 N, 0.92 kg Uniform white
PEG 600 60%
B PEG 14,000 40% 17 N, 1.73 kg 12 N, 1.22 kg Uniform white
PEG 600 60%
C PEG 1500 100% 30 N, 3.06 kg 27 N, 2.75 kg Opaque

Stability Tests RESULTS AND DISCUSSION


Stability studies were conducted over a 12-month pe- The final three suppositories selected for further char-
riod. Upon manufacture (time zero, T0 ), the supposito- acterisation were blend “A” (40% PEG 8000, 60% PEG
ries were stored under three different environmental 600), blend “B” (40% PEG 14000, 60% PEG 600) and
conditions to determine their stability; a refrigera- blend “C” (PEG 1500) (Table 1). There is a large vari-
tor at 4◦ C and 8% relative humidity (RH), a store ation in release onset in forms A, B and C, which is il-
containing a desiccator with saturated sodium hydro- lustrated by large standard deviation values. T100 for
gen sulphate solution at 20◦ C and 52% RH,20 and an PEG blends A, B and C were observed at 20, 45 and
Environmental Test Chamber (Copely, Nottingham, 60 min, respectively (n = 5–9). PEG blend C released
England) set at 30◦ C and 75% RH.21 These condi- the phenylalanine conjugate more slowly than PEG
tions were selected to simulate cold storage, typical blends A and B. This may be because of the greater
home storage and accelerated testing. As the sup- hardness of blend C (Table 2).
positories had low melting points, it was decided The fatty bases Witepsol (Gattefosse) and Gelucire
to reduce the commonly used accelerated tempera- (Sasol GmbH) produced a more homogenous blend
ture from 37◦ C to 30◦ C. The temperature in each of and melted instantly (data not shown), whereas the
the storage areas was monitored daily. DSC and in- PEG bases produced a more prolonged dissolution
frared (IR) spectroscopy were undertaken every week profile and dissolved more slowly over time (Fig. 2).
for 1 month, every month for 3 months and sub- It was decided to continue studies using PEG bases
sequently every 3 months for 12 months, and com- only, as they produced a more prolonged release of
pared with the T0 results.21 IR spectroscopy, was 20–60 min compared with 2 min for Witepsol (Gatte-
undertaken using a Nicolet IS10 IR from Thermo Sci- fosse) and Gelucire (Sasol GmbH). This slower disso-
entific (Fisher) with a Smart iTR attachment and lution of the PEGs allows the drug to be in contact
a diamond HATR (horizontal attenuated total re- with the rectum for longer where it is more likely
flectance). Thiol groups absorb at a wavenumber to be absorbed into the body. A sudden melt over
of 2600–2550 cm−1 , whereas disulphides absorb be- 2 min would increase the chances of the drug being
tween 620 and 600 cm−1 .22 These measurements were lost through expulsion.
used as the basis for the comparisons made between Figure 3 illustrates the release of the active from
samples. the three suppository bases over time. Cysteamine
Samples of uniformly less than 10 mg were placed hydrochloride was used as the active, formulated into
in a standard aluminium pan for DSC analysis. The suppository bases PEG blends A, B and C.
DSC was set to a heat–cool–heat cycle, where the sam- All three PEG blends containing cysteamine hy-
ple was equilibrated at 0◦ C, then heated to 100◦ C at drochloride as the active displayed more reproducible
10◦ C/min, equilibrated at 100◦ C, then cooled at 10◦ C/ profiles than dissolution using the phenylalanine con-
min to 0◦ C, before being heated again to 100◦ C at jugate. The PEG bases should dissolve at the same
10◦ C/min. rate in every replicate, and this may have been due

Table 2. Summary of Phenylalanine Conjugate Release Studies

Time to Release Phenylalanine Conjugate (Minutes, ±SD)

Percentage Release PEG Blend A PEG Blend B PEG Blend C Gelucire Witepsol
10% 2 (±7.1) 2 (±6.3) 3 (±5.1) 0.5 0.5
25% 5 5 9.5 1 1
50% 7 7.5 12.5 2 2
75% 10 (±5.6) 10 (±5.4) 17 (±8.6) 2.5 2.5
100% 20 (±0.6) 45 (±0.8) 59 (±0.7) 4.5 4.5

JOURNAL OF PHARMACEUTICAL SCIENCES DOI 10.1002/jps


SUPPOSITORY FORMULATIONS AS A POTENTIAL TREATMENT FOR NEPHROPATHIC CYSTINOSIS 5

Figure 2. Percentage release of the phenylalanine conjugate from PEG blends A, B and C
over time (n = 7, 9 and 6, respectively).

to the non-uniform drug loading of the phenylalanine of the suppository suggesting a uniform dispersion of
conjugate. The phenylalanine conjugate was devel- active within the PEG suppository.
oped initially to allow release from the dosage forms,
to be monitored spectrophotometrically. It should be Stability Tests
noted that cysteamine hydrochloride is a difficult The temperature and RH in each of the three storage
drug to formulate. At room temperature it undergoes chambers were monitored continuously throughout a
oxidation to a disulphide form, cystamine, which itself 12-month period. The average results obtained are
has been shown to deplete cells of cystine but which shown in Table 4.
is not licensed for use.5,23,24 Cysteamine is also ex-
tremely hygroscopic and deliquescent. However, the DSC Results: T0 , T6 Months, T12 Months Comparison
introduction of DTNB as a quantitative reagent al-
lowed measurement of cysteamine directly. A sum- The thermal properties of the suppositories were as-
mary of the release times obtained is shown in Table 3. sessed at T0 , 6 (T6 ) and 12 (T12 ) months storage at
The data produced using Ellman’s reagent demon- 4◦ C/8% RH, 21◦ C/52% RH and 30◦ C/75% RH. The
strated excellent reproducibility (average standard main melting endotherm of PEG in the broad range
error of the mean for PEG blend: A–0.39, B–0.58 and of 30◦ C–60◦ C was analysed in terms of the onset of
C–0.14). the melt (Tonset ), the peak temperature (Tmax ) and the
total enthalpy (W/g). Selected data are presented in
Table 5 as an increase or decrease in temperature and
enthalpy compared with samples at T0 . An exemplary
Active Dispersion Studies
thermogram of PEG blend C suppositories at T0 , T6
To ensure cysteamine hydrochloride was evenly dis- and T12 is also included (Fig. 5).
persed throughout the suppository, samples were Reference to the data in Table 5 indicates signifi-
taken from three separate portions of the suppository cant changes to Tonset and Tmax for aged samples of
and analysed using DSC (Fig. 4). There was minimal PEG blend A at 30◦ C, blend B at 21◦ C and blend C
difference between the tip, edge and middle sections at both 21◦ C and 30◦ C. Suppositories stored at 4◦ C

DOI 10.1002/jps JOURNAL OF PHARMACEUTICAL SCIENCES


6 BUCHAN ET AL.

Figure 3. Percentage release of cysteamine from PEG blends A, B and C over time (n = 6, 5
and 5, respectively).

demonstrated less certain variation in Tonset and Tmax a decrease in the molecular order of cysteamine con-
as may have been expected for samples stored at this taining PEG suppositories at elevated temperatures
refrigerated temperature. Although the bulk of the over time. Increased enthalpies for the melt transition
temperature variations were considered to be within in all samples are a consequence of a broadened en-
the error of measurement for DSC analyses, it is dotherm, symptomatic of increased disorder in crys-
clear that changes to the degree of crystallinity at the talline domains rather than increased order which
higher storage temperatures had occurred. A deple- would produce clear increases to values of Tonset and
tion in the crystalline character of PEG suppositories Tmax and a sharpening of the melting event. Such
may be expected to be based on the lengthy exposure changes are not evident.
to high humidity and temperature, exacerbated by the
IR Spectroscopy After 1 Week, 3 Months, 6 Months and
hygroscopic (deliquescent) nature of contained cys-
12 Months
teamine hydrochloride. There is some evidence of in-
creased crystallinity for PEG blend B stored at 21◦ C, Infrared spectroscopy utilises a set of unique peaks
considered as a `lamellar thickening” of ordered ethy- on a spectrum which can be used for identifying
lene oxide units,25 but the overall dataset suggests a compound. Each of the suppository samples was

Table 3. Summary of Cysteamine Hydrochloride Release Studies

Time to Release Cysteamine Hydrochloride (Minutes)

Percentage Release PEG Blend A PEG Blend B PEG Blend C


10% 1.75 2.5 1.75
25% 5 6.5 4.5
50% 9 10 7
75% 13 15 9.5
100% 26 26 17

JOURNAL OF PHARMACEUTICAL SCIENCES DOI 10.1002/jps


SUPPOSITORY FORMULATIONS AS A POTENTIAL TREATMENT FOR NEPHROPATHIC CYSTINOSIS 7

Figure 4. Section comparison of the melt phase between the tip, middle and edge areas of the
PEG blend A suppository containing cysteamine hydrochloride.

determined by IR spectroscopy and added to a com- ries at time zero. The results are shown in Table 6
pound library. A percentage match was then per- (selected examples only).
formed on each sample (Table 6). The stability tests using IR analysis demonstrated
The oxidation of cysteamine to cystamine was the that PEG blend C was the most stable in all con-
basis of comparisons made between samples. This re- ditions, with minimal changes over 6 months at
action produces a peak in the region of disulphide room temperature and accelerated tests, and up to
bond stretch, that is, 620–600 cm−1 , and allows a sim- 12 months stability when stored at 4◦ C. PEG blend
ple identification of sample degradation. B shows evidence of stability over 12 months at
Each suppository was analysed over time using the room temperature. PEG blend A shows evidence of
IR spectrometer, and compared with the supposito- degradation when subjected to a range of storage

Table 4. Average Temperatures and Relative Humidities in Each Storage


Chamber, With Standard Deviations (n = 3)

Refrigerator Store ETCa


Average temperature 3.7◦ C (±0.8) 20.3◦ C (±1.9) 29.9◦ C (±1.0)
Average relative humidity 9.2% (±0.3) 52% 76.4% (±2.0)
a Environmental Test Chamber.

Table 5. DSC Analysis Data of Selected Suppository Samples Over Time (n = 3)

T0 T6 Months T12 Months

Suppository Sample Tonset (◦ C) H (J/g) Tmax (◦ C) Tonset (◦ C) H (J/g) Tmax (◦ C) Tonset (◦ C) H (J/g) Tmax (◦ C)
PEG blend A, 4◦ C storage 50.6 71.8 56.4 +0.4 +4.2 +0.3 −0.9 +9.4 −1.3
PEG blend A, 21◦ C storage 50.7 83.8 57.7 +1.1 +9.0 +0.4 −0.2 +13.3 −0.5
PEG blend A, 30◦ C storage 50.5 75.3 56.6 −1.9 +8.8 −0.8 −5.9 +14.5 −5.1
PEG blend B, 4◦ C storage 51.4 73.4 57.8 +0.8 +7.3 −0.1 −0.3 +9.1 −1.3
PEG blend B, 21◦ C storage 50.4 67.2 56.7 +2.3 +19.2 +1.7 +0.6 +22.8 +0.2
PEG blend B, 30◦ C storage 50.7 80.1 57.6 +0.9 +6.1 −0.5 −0.8 +10.9 −1.7
PEG blend C, 4◦ C storage 41.2 159.4 50.3 +1.6 +20.4 −0.3 +0.9 +26.5 −0.2
PEG blend C, 21◦ C storage 41.1 165.9 50.3 −1.5 +6.5 −1.1 −3.5 +9.0 −2.9
PEG blend C, 30◦ C storage 50.0 164.5 40.9 −0.7 +6.1 −2.5 −15.3 −8.1 +5.3

DOI 10.1002/jps JOURNAL OF PHARMACEUTICAL SCIENCES


8 BUCHAN ET AL.

Figure 5. An Exemplary thermogram of PEG blend C supositories.

conditions, and therefore is unsuitable for the rectal CONCLUSIONS


delivery of cysteamine.
Liquid gel suppositories containing cysteamine have
previously been investigated for the treatment of
Correlation Between DSC Plots and IR Percentage
cystinosis.17 However, because of the osmotic nature
Match Results
of the suppository vehicle, the doses were rapidly
The IR percentage match data support the DSC plots. expelled and cysteamine blood plasma concentra-
This is evidence of correlation between the two meth- tions were insufficient to produce a reduction in
ods and allows a more conclusive result to be pro- mean leukocyte cystine concentration. In this project,
duced. For example, PEG blend C stored at 21◦ C dis- the suppository bases Witepsol (Gattefosse), Gelu-
played a 98.94% match at 6 months (Table 6). cire (Sasol GmbH) and PEG were investigated for
These stability test results correlate with the hard- their suitability as vehicles for the incorporation of
ness testing, and indicate that PEG blend C was cysteamine hydrochloride and a phenylalanine–cys-
the most stable formulation over a 12-month period. tamine conjugate. Melting point, hardness, stability
The suppositories stored at 4◦ C were generally more and appearance were analysed, and the three sup-
stable than those stored at higher temperatures, al- pository bases with the most suitable characteristics
though PEG blend C displays long-term stability even were chosen for further study. PEG blends A, B and
at room temperature. PEG blend B suppositories were C were made and characterised. Blend C displayed
stable over time at room temperature. PEG blend A good qualities (i.e., complete, reproducible release af-
suppositories displayed long-term instability under ter 30 min, stability over 12 months at 4◦ C/8% RH)
all storage conditions. PEG blend C displays ideal sta- required for the delivery of cysteamine hydrochlo-
bility over time in a range of storage conditions and ride to the rectum. DSC analysis indicated that cys-
is proposed as the optimal formulation in this study. teamine hydrochloride was dispersed uniformly in the

Table 6. Suppository Stability Over Time, Presented as Percentage Match to


Sample at T0 (Selected Data Only)

Suppository Blend/Storage Conditions Percentage Match to Sample at T0


PEG blend C, 4◦ C/8% RH 99.2% at 3 months 98.84% at 12 months
PEG blend A, 4◦ C/8% RH 97.4% at 3 weeks
PEG blend C, 21◦ C/52% RH 98.94% at 6 months
PEG blend B, 21◦ C/52% RH 97.95% at 12 months
PEG blend C, 30◦ C/75% RH 98.94% at 6 months

JOURNAL OF PHARMACEUTICAL SCIENCES DOI 10.1002/jps


SUPPOSITORY FORMULATIONS AS A POTENTIAL TREATMENT FOR NEPHROPATHIC CYSTINOSIS 9

suppositories. Dissolution studies using the pheny- 5. Gahl WA. 2009. Cystinosis. Pediatr Nephrol 6:1019–1038.
lalanine–cystamine conjugate revealed 20–60 min re- 6. Thoene JG. 2007. A review of the role of enhanced apopto-
lease, whereas cysteamine hydrochloride as the ac- sis in the pathophysiology of cystinosis. Mol Genet Metabol
92(4):292–298.
tive component demonstrated 17–26 min release on 7. Kleta R, Gahl WA. 2004. Pharmacological treatment of nephro-
average. The stability tests indicate that 4◦ C/8% RH pathic cystinosis with cysteamine. Expert Opin Pharmacother
provided the ideal storage conditions over a 12-month 5(11):2255–2262.
period. Formulation C was the most stable over time, 8. Hippert C, Dubois G, Morin C, Disson O, Ibanes S, Jacquet
C, Schwendener R, Antignac C, Kremer EJ, Kalatzis V.
whereas blend A was the least stable form, and even
2008. Gene transfer may be preventative but not curative
when stored in the refrigerator was subject to degra- for a lysosomal transport disorder. Mol Ther 16(8):1372–
dation. There was also evidence of an incompatibility 1381.
between cysteamine hydrochloride and PEG blends 9. Dohil R, Fidler M, Gangoiti JA, Kaskel F, Schneider
A and B, and this may be a combination of physical JA, Barshop BA. 2010. Twice-daily cysteamine bitartrate
therapy for children with cystinosis. J Pediatr 156(1):71–
ageing of the PEG and syneresis of cysteamine.26–29
75.
There was evidence that crystal ripening over time is 10. Schneider JA, Belldina EB, Huang MY, Brundage RC, Tracy
forcing the expulsion of the cysteamine hydrochloride TS. 2003. Steady-state pharmacokinetics and pharmacody-
to the outside surfaces of the suppository. The highly namics of cysteamine bitartrate in pediatric nephropathic
deliquescent cysteamine then quickly dissolves in en- cystinosis patients. Br J Clin Pharmacol 56:520–525.
11. Fidler MC, Barshop BA, Gangoiti JA, Deutsch R, Martin M,
vironmental moisture, forming droplets of liquid on
Schneider JA, Dohil R. 2006. Pharmacokinetics of cysteamine
the surface of the suppositories. For these reasons, bitartrate following gastrointestinal infusion. Br J Clin Phar-
PEG blends A and B would not be suitable for the macol 63(1):36–40.
rectal delivery of cysteamine. Analysis indicates that 12. Wenner WJ, Murphy JL. 1997. The effects of cysteamine on
PEG blend C would be an ideal suppository base for the upper gastrointestinal tract of children with cystinosis.
Pediatr Nephrol 11(5):600–603.
the delivery of cysteamine hydrochloride.
13. Schneider JA, Dohil R, Newbury RO, Sellers ZM, Deutsch R.
These tests demonstrate that cysteamine hy- 2003. The evaluation and treatment of gastrointestinal dis-
drochloride can be formulated as a suppository, and ease in children with cystinosis receiving cysteamine. J Pedi-
that the bases can tolerate varying amounts of drug atr 143(2):224–230.
loading. This will be of particular benefit when treat- 14. Bendel-Stenzel MR, Steinke J, Dohil R, Kim Y. 2008. Intra-
venous delivery of cysteamine for the treatment of cystinosis:
ing cystinosis during infancy, and should allow a sig-
Association with hepatotoxicity. Pediatr Nephrol 23:311–
nificant reduction in side effects, improving compli- 315.
ance and morbidity. In addition, suppositories may 15. Lim J, Pellois JP, Simanek EE. 2010. A retro-inverso TAT-like
help to eliminate the overnight treatment break peptide designed to deliver cysteamine to cells. Bioorg Med
which is difficult to overcome with the oral capsules. Chem Lett 20(21):6321–6323.
16. Walker WA. 2004. The stomach and duodenum. In Pediatric
Future work with the suppositories will involve the
gastrointestinal disease: Pathophysiology, diagnosis, manage-
development of in situ gelling forms, and should in- ment; Walker WA, Ed. Vol. 1. 4th ed. Shelton, Connecticut:
clude in vivo testing or an in vitro–in vivo correlation PMPH-USA, p 516.
to investigate the potential benefits these forms may 17. van’t Hoff W, Baker T, Dalton RN, Duke LC, Smith SP
have compared with the current oral treatment. Chantler C, Haycock GB. 1991. Effects of oral phosphocys-
teamine and rectal cyseamine in cystinosis. Arch Dis Child
ACKNOWLEDGMENTS 66:1434–1437.
18. Allen LV. 2008. Suppositories. 1st ed. London, UK: Pharma-
The authors gratefully acknowledge financial support ceutical Press.
from the Cystinosis Foundation, UK and TENOVUS, 19. Peppas NA, Sahlin JJ. 1989. A simple equation for the descrip-
Scotland. tion of solute release. III. Coupling of diffusion and relaxation.
Int J Pharm 57:169–172.
20. O’Brien FEM. 1948. The control of humidity by saturated salt
REFERENCES solutions. J Sci Instrum 25:73–76.
21. Aulton ME. 2002. Pharmaceutics. The science of dosage form
1. Almond PS, Matas AJ, Nakhleh RE, Morel P, Troppmann design. 2nd ed. London, UK: Churchill Livingstone.
C, Najarian JS, Chavers B. 1993. Renal transplantation 22. Coates J. 2000. Interpretation of infrared spectra, a practi-
for infantile cystinosis: Long-term follow-up. J Pediatr Surg cal approach. In Encyclopedia of analytical chemistry; Mey-
28(2):232–238. ers RA, Ed. Chichester, UK: John Wiley and Sons Ltd, pp
2. Gahl WA, Balog JZ, Kleta R. 2007. Nephropathic cystinosis in 10815–10837.
adults: Natural history and effects of oral cysteamine therapy. 23. Schneider JA, Thoene JG, Oshima RG, Crawhall JC, Olson DL.
Ann Intern Med 147(4):242–250. 1976. Cystinosis. Intracellular cystine depletion by aminoth-
3. Syres K, Harrison F, Tadlock M, Jester JV, Simpson J, Roy iols in vitro and in vivo. J Clin Invest 58:180–189.
S, Salomon DR, Cherqui S. 2009. Successful treatment of the 24. Skovby F, Kleta R, Anikster Y, Christensen R, Gahl WA. 2002.
murine model of cystinosis using bone marrow cell transplan- Cystinosis in Denmark: Mutation analysis and treatment with
tation. Blood 114(12):2542–2552. cystamine, the disulphide of cysteamine. Am J Human Genet
4. Cystinosis Research Network. 2007. About cystinosis. Lake 71(4):413.
Forest, Illinois: Cystinosis Research Network. Accessed May 25. Marand H, Huang Z. 2004. Isothermal lamellar thickening
20, 2007, at: www.cystinosis.org/symptoms-treatments.html. in linear polyethylene: Correlation between the evolution of

DOI 10.1002/jps JOURNAL OF PHARMACEUTICAL SCIENCES


10 BUCHAN ET AL.

the degree of crystalling and the melting temperature. Macro- 28. Talibuddin S, Runt J, Liu L, Chu B. 1998. Microstruc-
molecules 37:6492–6497. ture development and crystallization of poly(ethylene ox-
26. Zobrist B, Weers U, Koop T. 2003. Ice nucleation in aqueous ide) and melt-miscible PEO blends. Macromolecules 31:1627–
solutions of polyethylene glycol with different molar mass. J 1634.
Chem Phys 118(22):10254–10261. 29. Hay J. 1995. The physical ageing of amorphous and crys-
27. Li Y, Kaito A. 2006. Highly oriented structure formed in a talline polymers. Pure Appl Chem 67(11):1855–1858. Accessed
lamella-forming diblock copolymer with high molar mass. Eur 02/02/2012, at: http://iupac.org/publications/pac/pdf/1995/pdf/
Polym J 42:1986–1993. 6711x1855.pdf.

JOURNAL OF PHARMACEUTICAL SCIENCES DOI 10.1002/jps

You might also like