throbber

`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`DOI:10.1111/j.1365-2125.2005.02517.x
`
`British Journal of Clinical Pharmacology
`
`Gastrointestinal transit, release and plasma
`pharmacokinetics of a new oral budesonide formulation
`
`1
`1
`2
`1
`3
`4
`5
`5
`5
`M. Brunner,
` S. Ziegler,
` A. F. D. Di Stefano,
` P. Dehghanyar,
` K. Kletter,
` M. Tschurlovits,
` R. Villa,
` R. Bozzella,
` G. Celasco,
`
`5
`2
`3
`1
`L. Moro,
` A. Rusca,
` R. Dudczak
` & M. Müller
`1
`3
`Department of Clinical Pharmacology, Division of Clinical Pharmacokinetics,
`Department of Nuclear Medicine, Medical University of
`2
`4
`5
`Vienna, Vienna, Austria;
`Cross Research S.A., Arzo, Switzerland;
`Atomic Institute of the Austrian Universities, Vienna, Austria; and
`Cosmo
`S.p.A., Lainate, Italy
`
`Correspondence
`Martin Brunner, MD,
` Department of
`Clinical Pharmacology, Medical
`University of Vienna, Allgemeines
`Krankenhaus – AKH,
`Waehringer-Guertel 18–20, A-1090
`Vienna, Austria.
`+
`Tel:
`
`43 140400 2981
`+
`Fax:
`
`43 140400 2998
`E-mail:
`
`martin.brunner@meduniwien.ac.at
`
`Keywords
`budesonide, gastrointestinal transit,
`targeted drug delivery, pharmaco-
`scintigraphy, food effect
`
`Received
`28 February 2005
`Accepted
`16 June 2005
`
`Aims
`The aims of the study were to: (1) evaluate the gastrointestinal transit, release and
`absorption of budesonide from tablets with a new multimatrix formulation (M MX®)
`designed to release the drug throughout the whole colon, and (2) assess the
`influence of food on budesonide bioavailability.
`
`Methods
`Two phase I studies, each comprising 12 healthy males, were performed. Gastrointes-
`153
`Sm-labelled tablets containing 9 mg budesonide was evaluated by
`tinal transit of
`means of pharmaco-scintigraphy. The effect of food was tested by comparing plasma
`pharmacokinetics after intake of a high fat and high calorie breakfast with fasting
`controls.
`
`Results


`153
` 6.9 h.
` SD 9.8
`Sm-labelled tablets reached the ascending colon after a mean
`Initial tablet disintegration was observed in the ileum in 42% and the ascending and
`transverse colon in 33% of subjects. Ninety-six per cent of the dose was absorbed
`into the systemic circulation during passage through the whole colon including

`C
` values from 1429
` 1014 to
`the sigmoid. Food significantly decreased
`max

`=

`−
`1
`P
` 601 pg mL
` (
`
` 0.028) and AUC values
`from 14 814
` 11 254
`to
`1040

`=
`−
`−
`1
`1
`P
`t
` 9369 pg h
` mL
` (
`
` 0.008). Mean residence time and
` increased by
`13 486
`max
`12–29%. There was no drug accumulation after 1 week of once daily oral adminis-
`tration of budesomide.
`
`Conclusions
`MMX®-budesonide tablets appear suitable for targeted colonic drug delivery. Transit
`parameters and low systemic bioavailability warrant further studies with the new
`formulation.
`
`Introduction
`The pharmacological treatment of inflammatory bowel
`diseases (IBD) is determined by the location, extent and
`severity of the disease within the gastrointestinal tract.
`The drugs include aminosalicylate formulations, corti-
`costeroids, antibiotics, immunomodulators, and mono-
`
`clonal antibodies [1, 2]. Although corticosteroids are an
`effective option for disease management, dose- and
`duration-dependent side-effects might limit their long-
`term use. Budesonide, a nonhalogenated glucocorticos-


`, 17-butylidendioxy-11
`, 21-dihydroxy-1,4-
`teroid (16
`pregnadien-3,20-dione), is part of a group of new topical
`
`© 2005 Blackwell Publishing Ltd
`
`Br J Clin Pharmacol
`
`61
`
`:1
`
`31–38
`
`31
`
`Cosmo Ex 2001-p 1
`Mylan v Cosmo
`IPR2017-01035
`
`

`

`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`M. Brunner et al.
`
`corticosteroids, characterized by potent local anti-
`inflammatory activity, and was initially introduced for
`the treatment of asthma and rhinitis. Due to an extensive
`first-pass elimination its systemic bioavailability is only
`10–15% compared with other corticosteroid formula-
`tions, thus, improved safety and tolerability might be
`anticipated [2].
`For the treatment of IBD, budesonide has been eval-
`uated either as oral controlled-ileal-release formulation
`targeting the distal ileal and right-sided colonic region
`in Crohn’s disease [3], or as enema for the treatment of
`left-sided ulcerative colitis or sigmoiditis [4]. However,
`there are no budesonide formulations available for the
`oral treatment of distally located IBD. To allow the
`homogenous release of budesonide along the whole
`colon at a controlled rate, new gastro-resistant, extended
`release tablets characterized by a multimatrix structure
`(i.e. MMX®-tablets containing 9 mg budesonide), have
`been developed.
` performance of such novel
`To evaluate the
`in vivo
`delivery systems, the noninvasive technique of gamma-
`scintigraphy is routinely employed [5]. This technique
`allows monitoring of the gastrointestinal-transit of
`orally ingested dosage forms, to identify the exact time
`and region of disintegration and to follow the release of
`the active ingredient. Consequently, it is possible to
`relate the plasma and urine pharmacokinetics of the drug
`to the scintigraphic pattern within the gastrointestinal-
`tract and to determine the rate and extent of absorption
`in a defined region of interest (a process termed
`‘pharmaco-scintigraphy’). In the present study, budes-
`onide tablets were labelled by the addition of a non-
`radioactive tracer, which is not absorbed from the
`gastrointestinal-tract, namely
`samarium-152-oxide,
`153
`Sm) by neu-
`which was converted to samarium-153 (
`tron activation before tablet administration [6].
`The present paper describes the results of two inde-
`pendent studies with MMX®-tablets in healthy subjects
`designed to (1) evaluate the gastrointestinal-transit,
`release and absorption of budesonide from MMX®-
`tablets using pharmaco-scintigraphy; (2) assess the
`influence of food on the bioavailability of budesonide;
`(3) characterize the steady-state pharmacokinetics of
`budesonide, and (4) evaluate safety and tolerability of
`the new MMX®-budesonide formulation after a 1-
`week, once daily treatment regimen.
`
`Subjects and methods
`Study design
`Two independent, phase I studies were performed in two
`different study populations. One was a single dose phar-
`maco-scintigraphic pilot study and the other an open,
`
`32
`
`61
`
`:1
`
`Br J Clin Pharmacol
`
`randomized, balanced, single and multiple dose pharma-
`cokinetic study. Both were approved by the local Ethics
`Committees and were performed in accordance with the
`Declaration of Helsinki and the Good Clinical Practice
`Guideline of the European Commission (EC-GCP
`guideline). All subjects received a detailed description
`of the study and written informed consent was obtained.
`
`Study populations
`Twelve male healthy Caucasian subjects took part in
`each study. In the first, the mean age of the subjects was


`32
` 5 years, mean height 178
` 6 cm, mean weight


`−
`2
`. In the
`81.1
` 12.5 kg, and mean BMI 25.5
` 3.1 kg m

`second study the mean age was 22
` 4 years, mean


`height 177
` 8 cm, mean weight 74.1
` 9.2 kg, and BMI

`−
`2
`.
`23.5
` 2.6 kg m
`Before start of each study, subjects were evaluated by
`medical history, physical examination, 12-lead electro-
`cardiogram, measurements of blood pressure and heart
`rate, complete haematology with differential white
`blood cell count, blood chemistry, hepatitis B surface
`antigen, hepatitis C antibody and HIV antibody tests,
`urinalysis and urine drug screening. Subjects were
`excluded if they had taken any prescribed medication or
`over-the-counter drugs within a period of 2 weeks
`before the study. Subjects were excluded from the phar-
`maco-scintography if they had undergone any diagnos-
`tic analysis with radioactive tracers or X-rays during the
`6 months preceding the study.
`
`Study medication
`The study medication for both studies was provided by
`Cosmo S.p.A., Lainate (MI), Italy, and consisted of
`round, film-coated, gastro-resistant, extended-release
`tablets, with multimatrix structure (MMX®) [7], a
`diameter of 10 mm, a weight of 330 mg, and each con-
`taining 9 mg budesonide. Tablets were designed for
`slow and graded budesonide release in the colon, and
`consisted of an inner lipophilic matrix in which the
`active ingredient was dispersed, an outer hydrophilic
`matrix generated by
` hydration of selected poly-
`in situ
`mer chains and a third amphiphilic matrix promoting the
`inert matrix wettability. Tablets were film-coated with
`polymethacrylate to provide gastro-resistance.
`152
`Sm
`O
` (1.67% w/w per
`For scintigraphy, 5 mg of
`2
`3
`152
`tablet) was added to each tablet. Stable
`Sm-oxide was

`-ray-
`subsequently transformed into the radioactive,
`153
`Sm isotope by neutron activation. Before the
`emitting
`start of the study, preliminary tests were performed on
`152
`Sm
`O
` tablets to determine the most optimal condi-
`2
`3
`tions for activation. Tablets were irradiated for different
`time periods (1–10 min) under different neutron fluxes
`
`Cosmo Ex 2001-p 2
`Mylan v Cosmo
`IPR2017-01035
`
`

`

`−
`−
`−
`11
`13
`2
`1
`) to obtain the intended
` s
` neutrons cm
`10
`(10
`radioactivity level of 0.8 MBq/tablet (i.e. 0.16 MBq/mg
`Sm
`O
`) at the time of drug administration.
` dis-
`In vitro
`2
`3
`solution tests were performed to verify that the release
`profiles of the tablets were not significantly altered by
`the irradiation procedure. After irradiation, the budes-
`onide content of the tablet was determined to verify that
`no degradation of the drug had occurred. Neutron acti-
`vation was found not to alter the pharmaceutical prop-
`erties and the analytical purity of the formulation. The

`mean (
`SD) amount of radioactivity administered was

`1.1
` 0.4 MBq/dose, which is in compliance with the
`Council Directive 96/29 EURATOM, and with the gen-
`eral guidelines of the World Health Organization.
`
`Experimental design
`Subjects attended the Clinical Trial Center in the
`evening before drug administration and remained under
`observation for 24 h postdose. During their stay, sub-
`jects received standardized meals according to normal
`caloric needs for adult healthy males of normal weight
`with slight physical activity. After an overnight fast, the
`study medication was administered orally between
`07.00 and 08.00 h with a defined amount of water.
`Thereafter, subjects underwent scintigraphic scans,
`blood and urine sampling at predetermined intervals up
`to 24 h postdose. To improve scan interpretation, each
`subject had four radioactive point sources taped to their
`skin in the following anatomical sites: the lower end of
`the sternum, the umbilicus, and the left and right iliac
`spine. The transit of budesonide tablets along the gas-
`trointestinal-tract was recorded with the subjects sitting

`under a large field of view, double-head
`-camera (Axis,
`Picker®) equipped with a low-energy, all-purpose, par-
`allel-hole collimator. Scanning was performed at 3 min
`postdose, and at approximately 20-min intervals up to
`3 h, and then at 30-min intervals up to 10 h. Additional
`scans were taken at 12 and 24 h postdose. In each sub-
`ject, the following regions of interest (ROIs) were iden-
`tified: stomach, small intestine, terminal ileum-caecum,
`ascending colon, transverse colon, descending colon,
`sigmoid colon. Data were stored electronically. The
`location of the labelled formulation in the gastrointesti-
`nal-tract was established by viewing the image on a
`monitor. Quantitative data were obtained by measuring
`the count rates recorded from the ROIs. The geometric
`mean values of the corresponding anterior and posterior
`count rates were calculated and corrected for radioactive
`decay. The appearance or disappearance of the labelled
`formulation to or from the ROIs was evaluated by
`recording the time of the first and last appearance of
`radioactivity in the region.
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`Gastrointestinal budesonide transit
`
`Venous blood samples (10 mL) were taken from an
`arm vein at the following times: 0 (predose), 1, 2, 3, 3.5,
`4, 4.5, 5, 6, 7, 8, 10, 12, and 24 h. Plasma samples,
`≤
`−

`obtained by centrifugation were stored at
`
`20
`C until
`analysis.
`
`Single-dose pharmacokinetic and effect of food study
`Subjects attended the Clinical Trial Centre in the
`evening before drug administration and received a stan-
`dardized dinner between 20.00 and 21.00 h Subjects
`were randomized into two groups of six. In the morning,
`one group received a high fat, high caloric breakfast, i.e.
`1000 kcal with fat accounting for 50% of the total
`caloric content. One MMX®-budesonide tablet was
`administered within 30 min after the start of breakfast
`with a defined amount of water. The other group
`received one MMX®-budesonide tablet after an over-
`night fast of at least 10 h. All subjects were discharged
`on the third day and returned to the Clinical Trial Centre
`after a 7-day wash-out period for the second phase of
`the cross-over study.
`
`Multiple–dose pharmacokinetic study
`After a 7-day wash-out period the 12 subjects who took
`part in the single dose study attended the trial centre and
`remained confined for 8 consecutive nights. One
`MMX®-budesonide tablet was administered once daily
`in the morning after an overnight fast of 12 h for 7
`consecutive days. Subjects were discharged on day 8.
`Venous blood samples (10 mL) were collected from
`an arm vein at predose on days 2, 4, 6 and 7, and at the
`following time points after the last dose on the 7th
`treatment day: 0 (predose), 2, 3, 4, 5, 6, 7, 8, 10, 12, 16,
`20, 24, 30, 36 and 48 h. Plasma samples, obtained by
`≤
`−

`centrifugation were stored at
`
`20
`C until analysis.
`
`Clinical assessment
`The nature, severity and frequency of adverse events,
`laboratory values outside the normal range and abnor-
`mal ECG measurements were documented.
`
`Budesonide analysis
`All plasma samples were analysed for their budesonide
`content at Pharmakin GmbH, Ulm, Germany using a
`validated GC-MS/NCI method with SIM-detection. Fifty

`L of 2
`microlitres of triamcinolone acetonide and 50
`M
`NaOH were added to previously thawed and homoge-
`nized 1 mL plasma samples. Budesonide was extracted
`into n-pentane: dichloromethane (70 : 30 v/v). After cen-
`trifugation the organic layer was evaporated to dryness

`C. For derivatization
`under a stream of nitrogen at 40

`L mixture of 12.5% acetic anhydride/12.5% tri-
`a 100-
`
`Br J Clin Pharmacol
`
`61
`
`:1
`
`33
`
`Cosmo Ex 2001-p 3
`Mylan v Cosmo
`IPR2017-01035
`
`

`

`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`M. Brunner et al.
`
`ethylamine in acetonitrile was added to the dry residue.
`After a reaction time of 15 min at room temperature, the
`mixture was dried under nitrogen stream and the residue

`reconstituted in 30
`L of ethyl acetate. Two microlitres
`of the derivatized extract were subjected to GC-MS anal-
`ysis. The gas chromatograph was equipped with fused
`silica capillary column for the separation of budesonide
`and internal standard triamcinolone acetonide and cou-
`pled to a mass spectrometer. The carrier gas was helium
`−
`1
`. The GC–MS interface
`5.0 at a flow rate of 2.6 mL min

`was at a pressure of 10.1 psi and a temperature of 285
`C.
`The negative chemical ionization mode was used with
`methane 3.5 as ionization gas. The lower limit of quan-
`−
`1
`. Precision
`tification (LLQ) of the assay was 50.0 pg mL
`at the LLQ expressed as a coefficient of variation was
`6% for the samples from both studies.
`
`Data analysis
`In the pharmaco-scintigraphic study the relative percent-
`age of drug absorption in the time during which the
`radioactivity was detectable in the target region (i.e.
`between the ascending and the descending colon), was
`taken as primary outcome parameter and was calculated
`by means of the following equation: Relative percentage
`=

`/AUC
`), where AUC
` is
`absorption
` 100
` (AUC
`target
`24
`target
`the area under the plasma
` time curve in the target
`vs.
` the area under the plasma concentra-
`region and AUC
`24
` time curve for drug up to 24 h postdosing. The
`tion
`vs.
`following variables were described: (1) gastric emptying
`time; (2) small intestinal transit; (3) ileal transit; (4)
`colonic transit; (5) time of initial tablet disintegration.
`Measurement of the distribution of radioactivity was
`achieved by determining the count rates recorded from
`the ROIs. Geometric means of corresponding anterior
`and posterior count rates were calculated and corrected
`for radioactive decay. Whenever plasma samples were
`missing at the start and end times of transit in the rele-
`vant regions, plasma concentrations were obtained by
`linear interpolation of the concentrations available at the
`times immediately preceding and following the time of
`interest.
`For both studies the pharmacokinetic parameters for
`budesonide were calculated using Kinetica Software,
`Version 2000 (Innaphase Corporation, Philadelphia, PA,
`USA).
`
`Statistical analysis

`Mean
`SD data were calculated using SAS® software
`version 8.2 for Windows® (SAS Institute Inc., USA).
`Statistical comparisons of pharmacokinetic data were
`performed using Kinetica Software. A P-value <0.05
`was considered to be statistically significant.
`
`34
`
`61
`
`:1
`
`Br J Clin Pharmacol
`
`Cmax values were compared using the analysis of vari-
`ance (ANOVA) at the level of significance of P < 0.05
`with study treatment (single dose vs. multiple dose) as
`covariate and the 90% CI for log-transformed data. The
`coefficient of accumulation after repeated administra-
`tion was calculated as the repeated/single dose ratios for
`Cmax and AUC.
`According to the latest version of an FDA guideline
`on Food-Effect Bioavailability and Fed Bioequivalence
`Studies [8], ‘an absence of food effect on BA is not
`established if the 90% CI for the ratio of population
`geometric means between fed and fasted treatments,
`based on log-transformed data, is not contained in the
`equivalence limits of 80–125% of either AUC∞ (AUC48
`when appropriate) or Cmax’. Thus, AUC and Cmax calcu-
`lated for the two diet regimens (fed and fasted) after
`administration were compared by analysis of variance
`(ANOVA) for a cross-over design (log-transformed data)
`at the level of significance P < 0.05. The 90% CI for the
`ratio of population geometric means between fed and
`fasted condition was calculated. tmax after administration
`of drug under fed or fasted conditions was compared
`using the nonparametric Friedman test (nontransformed
`data).
`
`Results
`MMX®-budesonide tablets were detected by scinto-
`graphic imaging in the ascending colon between 4 and
`>24 h after dosing (Figure 1). The drug left the descend-
`ing colon at 12 to >24 h postdosing. To estimate the
`relative percentage of budesonide absorbed from the
`target region (i.e. that between the ascending and
`the descending–sigmoid colon) the AUCtarget/AUC24
`ratio was calculated from the plasma AUC over the time
`during which radioactivity was detectable in the target
`region (mean ± SD AUCtarget: 15 114 ± 14 402 pg
`h−1 mL−1) and plasma AUC values during the 24 h obser-
`vation period (mean AUC24: 15 607 ± 14 549 pg h−
`1 mL−1). The mean relative absorption was 95.9 ± 4.2%
`indicating that during the study period absorption of
`budesonide occurred throughout the whole colon
`including the sigmoid.
`Initial tablet disintegration/erosion (ITD) started at
`9.48 ± 5.11 h after administration either in the small
`intestine (n = 2), the ileum (n = 5), the ascending
`(n = 2), transverse (n = 2) or sigmoid colon (n = 1). Indi-
`vidual times and location are given in Table 1. The times
`of tablet residence in different ROIs were 17–117 min
`(stomach), 37 min to 9.95 h (small intestine), 0.5–12 h
`(ileum), 1.5 to >15.5 h (ascending colon), 2 to >17 h
`(transverse colon), and 12 to >17 h (descending colon).
`Due to the absence of scans between 12 and 24 h after
`
`Cosmo Ex 2001-p 4
`Mylan v Cosmo
`IPR2017-01035
`
`

`

`Table 1
`Individual times and locations of initial tablet disintegration
`(ITD) within the gastrointestinal tract of 153Sm-labelled
`MMX®-budesonide tablets
`
`Subject
`
`ITD (h)
`
`Location in the gastrointestinal tract
`
`01
`02
`03
`04
`05
`06
`07
`08
`09
`10
`11
`12
`
`>24
`8.5–9
`12–24
`10–12
`4–4.5
`10–12
`5.5–6
`7–7.5
`6–6.5
`8–8.5
`9.5–10
`6.5–7
`
`Sigmoid colon
`Ascending colon
`Ileum/ascending colon
`Small intestine/ileum
`Ileum/ascending colon
`Ileum
`Transverse colon
`Small intestine/ileum
`Ileum
`Ileum/ascending colon
`Transverse/descending colon
`Ascending/transverse colon
`
`Gastrointestinal budesonide transit
`
`high
`
`low
`
`Figure 1
`A representative scintigraphic image, depicting the dispersion of 153Sm-
`labelled MMX®-budesonide tablets in the colon. The image shown was
`acquired approximately 7 h after drug administration
`
`3000
`
`STOMACH
`
`SMALL INTESTINE
`
`ILEUM
`
`ASCENDING COLON
`TRANSVERSE COLON
`DESCENDING COLON
`SIGMOID COLON
`
`2500
`
`2000
`
`1500
`
`1000
`
`500
`
`0
`
`0
`
`Budesonide plasma concentration (pg/mL)
`
`Figure 2
`The mean ±SD plasma concentration vs. time
`profile of budesonide after single-dose
`administration of 153Sm-labelled MMX®-tablets
`to 12 healthy males. Lines depict periods
`between minimal time to arrive and maximal
`time to leave different gastrointestinal regions
`
`3
`
`6
`
`9
`
`15
`12
`Time (hrs)
`
`18
`
`21
`
`24
`
`drug administration, transit times in the transverse,
`descending and sigmoid colon and the percentage of
`drug absorption in the target ROI were approximated or
`not available.
`Budesonide was first detected in plasma 6.8 ± 3.2 h
`post administration (tlag) (Figure 2). The mean time to
`reach Cmax (tmax) was 14.0 ± 7.7 h and the mean Cmax of
`1768.7 ± 1499.8 pg mL−1. The difference between tmax
`and tlag was 7.2 ± 5.5 h. This period may be regarded as
`
`the time during which most of the drug is released and
`absorption dominates over elimination.
`Following oral single dose administration, budes-
`onide was detectable in plasma between 3 and 16 h
`under fasting conditions and between 5 and 16 h after a
`meal. The corresponding peak concentrations occurred
`between 12 and 24 h and between 10 and 36 h. Mean
`budesonide plasma concentration vs. time profiles after
`single dose administration under fasting and fed condi-
`
`Br J Clin Pharmacol
`
`61:1
`
`35
`
`Cosmo Ex 2001-p 5
`Mylan v Cosmo
`IPR2017-01035
`
`

`

`M. Brunner et al.
`
`Table 2
`The plasma pharmacokinetics of budesonide after a single
`oral dose of MMX®-budesonide tablets containing 9 mg
`of the drug to fasting and fed subjects. Results are
`presented as means ±SD, n = 12
`
`Table 3
`The plasma pharmacokinetics of budesonide at steady-
`state following once daily administration of MMX®-
`budesonide tablets containing 9 mg of the drug for 7 days.
`Results are presented as means ±SD, n = 12
`
`Fasting
`
`Fed
`
`Mean ±SD
`
`Max
`
`Min
`
`CV%
`
`Cmax (pg mL−1)
`tlag (h)
`tmax (h)
`AUC48 (pg h −1 mL−1)
`AUC∞ (pg h −1 mL−1)
`thalf (h)
`MRT (h)
`
`1428.7 ± 1013.5
`7.4 ± 4.2
`16.0 ± 3.4
`14 814 ± 11254
`15 503 ± 11340
`5.4 ± 2.0
`19.9 ± 4.6
`
`1039.9 ± 601.4
`9.8 ± 3.6
`20.7 ± 8.7
`13 486 ± 9369
`12 512 ± 7569*
`5.6 ± 2.9*
`22.3 ± 6.0*
`
`*Mean and SD of 10 subjects; Cmax, maximum plasma
`concentration; tlag, time to detect drug concentration in
`plasma; tmax, time to achieve Cmax; AUC48, area under
`the concentration curve from administration to last
`observed; concentration time t (48 h).; AUC∞, area under
`the concentration/time curve extrapolated to infinity; thalf,
`elimination half-life; MRT, mean residence time.
`
`Cssmin
`Cssmax
`tssmax
`Caverage
`AUCss
`%PTF
`
`109.9 ± 75.3
`891.3 ± 294.1
`11 ± 4.9
`387.3 ± 153.9
`9295.2 ± 3694.2
`205.9 ± 83.9
`
`269.4
`1433.5
`16
`607.8
`14588
`412.5
`
`0
`158
`0
`82.4
`1978.5
`128.5
`
`68.5
`44.2
`44.9
`39.7
`39.7
`40.7
`
`Cssmin, minimum plasma concentration at steady state;
`Cssmax , maximum plasma concentration at steady state;
`tssmax, time at which Cssmax is achieved; Caverage, mean or
`average steady state drug concentration; AUCss, area
`under the concentration/time curve during the selected
`dosing; interval at steady state calculated with trapezoidal
`method; %PTF, peak-through- fluctuation percentage.
`
`68–134% for Cmax and 67–131% for AUC48. Mean resi-
`dence time (MRT) and tmax were increased by 12–29%
`after a meal (Table 2), compared with fasting, but the
`difference was not statistically significant (Friedman
`P = 0.248).
`Twenty-four hours after the first dose of the dose
`regimen, plasma concentrations ranged from 105 to
`1163 pg mL−1 Pre-dose concentrations on days 4 and
`6 were 409 ± 379 pg mL−1 and 336 ± 259 pg mL−1,
`respectively. After the final dose in the morning of day
`7 budesonide concentrations reached peak concentra-
`tions after 11 ± 4.9 h. Forty-eight hours after the last
`dose, all plasma budesonide concentrations were below
`the limit of detection. Table 3 summarizes the pharma-
`cokinetic parameters at steady state. The AUCss/AUC∞
`and Cssmax/Cmax ratios were 0.82 ± 0.47 (90% CI: 47.88–
`103.44%) and 0.87 ± 0.51 (90% CI: 46.58–97.71%),
`indicating a lack of drug accumulation after the 1-week
`treatment period.
`During both studies no serious adverse events were
`reported. The study medication was well tolerated by all
`subjects.
`
`Discussion
`In the treatment of patients with moderately severe
`ulcerative colitis or Crohn’s disease, oral corticosteroids
`such as prednisolone are commonly administered when
`5-aminosalicylate-based compounds are not effective.
`
`2500
`
`2000
`
`1500
`
`1000
`
`500
`
`0
`
`0
`
`6
`
`12
`
`30
`24
`18
`Time (hrs)
`
`36
`
`42
`
`48
`
`Budesonide plasma concentration (pg/mL)
`
`Figure 3
`The mean ±SD plasma concentration vs. time profiles of budesonide after
`single-dose administration of 9 mg MMX®-tablets to 12 males under
`fasting (䉭) and fed conditions (䉮)
`
`tions are depicted in Figure 3, and the pharmacokinetic
`data are summarized in Table 3.
`Food significantly decreased Cmax and AUC48
`(P = 0.028 and P = 0.008, respectively). The 90% CI
`values for the percentage ratio of population geometric
`mean values between fed and fasted conditions were
`
`36
`
`61:1
`
`Br J Clin Pharmacol
`
`Cosmo Ex 2001-p 6
`Mylan v Cosmo
`IPR2017-01035
`
`

`

`However, the potent anti-inflammatory activity of corti-
`costeroids must be balanced against potential, some-
`times serious side-effects [1].
`Owing to its extensive hepatic first pass metabolism
`budesonide is likely to cause less steroid related side-
`effects [9]. Currently, oral budesonide is available as
`controlled ileal release formulation for targeted drug
`delivery to inflamed intestinal regions. This time- and
`pH-dependent delivery system consists of enteric coated
`(Eudragit L) pellets with a rate-limiting polymer con-
`taining the active drug. This formulation releases 70%
`of budesonide in the distal ileum and right sided colon
`[3], the main location of Crohn’s disease lesions. In the
`present study, the release, absorption and plasma phar-
`macokinetics of budesonide were evaluated after admin-
`istration of newly patented MMX®-tablets to healthy
`subjects. The MMX® matrix has a polymeric structure
`and was designed to slowly and homogenously release
`the active drug at a controlled rate throughout the whole
`colon for the oral treatment of IBD with a more distal
`location than the ileo-caecal region. This formulation
`has been successfully employed for the delivery of ami-
`nosalicylate [7].
`Gastrointestinal-transit was studied by means of
`pharmaco-scintigraphy. Evaluation of the scintigraphic
`images showed that 153Sm-labelled MMX®-budesonide
`tablets reached the colonic region after a mean of 9.8 h.
`Initial tablet disintegration was observed in the ileum in
`42% of subjects, whereas in 33% the main site of dis-
`integration was either the ascending or transverse colon.
`Budesonide plasma concentrations were first detected
`after 6.8 ± 3.2 h, whereas maximum plasma concentra-
`tions were reached approximately 7 h
`later,
`i.e.
`14.0 ± 7.7 h after drug administration. These findings
`suggest that drug release began before break-down of
`the tablet matrix. The lag-time between the initial detec-
`tion of budesonide in plasma and tmax indicates sustained
`drug release from MMX®-tablets. Furthermore, about
`96% of budesonide absorption took place during the
`time tablets were passing the region between the ascend-
`ing and the descending/sigmoid colon, which underlines
`the efficiency of the colon targeting release kinetics of
`the new MMX® matrix structure.
`The systemic availability of budesonide from
`MMX®-tablets matches that of marketed formulations
`[3]. However, the time to reach maximum plasma con-
`centrations for the MMX®-tablets was prolonged from
`6 h [10] to 14 h, which might be a measure of the pro-
`jected slow and graded budesonide release throughout
`the colon. Furthermore, as reported previously for other
`formulations [11], the gastrointestinal-transit and sys-
`temic absorption of budesonide from MMX®-tablets
`
`Gastrointestinal budesonide transit
`
`were subject to a high interindividual variability with
`times of arrival in and times of leaving from some ROIs
`differing up to 10-fold between subjects.
`One factor, that might contribute to variable gas-
`trointestinal-transit times, is concurrent food and drug
`intake [12], which, together with tablet size, is known
`to affect gastric emptying. Tablets of 10 mm diameter
`or less empty from the fed stomach in a linear fashion
`[5], whereas an increase in tablet size and concomitant
`food intake lead to increased variability in gastric
`emptying. To test the effect of food on budesonide phar-
`macokinetics, plasma samples were taken after admin-
`istration of a high calorie, high fat breakfast according
`to FDA guidelines [8]. Food intake significantly
`decreased the rate and extent of budesonide absorption.
`MRT and tmax increased by up to 30% compared with
`fasted control subjects. Delayed gastric emptying might
`explain the observed retarded postprandial absorption of
`budesonide, a finding, which has also been described for
`controlled ileal budesonide formulations [13]. In turn,
`decreased plasma AUC and Cmax, may either result from
`a food–drug interaction affecting bioavailability or from
`increased presystemic metabolism due to postprandial
`changes in liver blood flow, which might affect drugs
`with high intrinsic hepatic clearance such as budes-
`onide. The significant decrease in systemic exposure,
`which may improve the safety profile of the drug, could
`justify the administration of MMX®-budesonide with a
`meal. Furthermore, the lack of drug accumulation and
`serious side-effects after a 7-day treatment period sup-
`ports the long-term treatment with the new drug formu-
`lation. The duration of remission in Crohn’s disease
`patients has recently been linked to the length of corti-
`costeroid treatment [14].
`The present study was performed in healthy subjects
`and disease activity and the possibility that disease
`activity and inflammation might affect gastrointestinal-
`transit and drug absorption should be considered.
`Although previous studies on the uptake of budesonide
`in the ileo-colonic region have shown no major differ-
`ences between patients and healthy subjects, a disease
`mediated influence on transit times should not be com-
`pletely ruled out [3]. In particular, inflammation has
`been reported to significantly affect colon transit, such
`that ascending colon transit shortens with more drug
`remaining to be delivered to and absorbed by the distal
`colon [15]. To what extent inflammation influences
`mucosal drug absorption of budesonide is currently a
`matter of debate, as systemic exposure in patients with
`active Crohn’s disease has been described to be either
`increased or decreased compared with healthy controls
`[10, 13]. Besides inflammation, the absorption of budes-
`
`Br J Clin Pharmacol
`
`61:1
`
`37
`
`Cosmo Ex 2001-p 7
`Mylan v Cosmo
`IPR2017-01035
`
`

`

`M. Brunner et al.
`
`onide might also be affected by the expression of P-
`glycoprotein, an intestinal efflux pump, which actively
`secretes substrates into the gut lumen. Recently, budes-
`onide has been identified as a P-glycoprotein substrate
`and poor response to corticosteroid treatment in IBD has
`been related to increased efflux pump expression [16,
`17].
`In conclusion, MMX®-budesonide tablets appear
`suitable for targeted drug delivery to the colon. Transit
`parameters, low systemic bioavailability and a lack of
`significant side-effects after repeated administration
`support further studies on the effectiveness of this new
`formulation of budesonide in controlled clinical trials in
`IBD patients.
`
`Competing interests: None declared.
`
`References
`1 Podolsky DK. Inflammatory bowel disease. N Engl J Med 2002;
`347 (6): 417–29.
`2 Navarro F, Hanauer SB. Treatment of inflammatory bowel disease:
`safety and tolerability issues. Am J Gastroenterol 2003; 98
`(12(Suppl): S18–23.
`3 Edsbäcker S, Andersson T. Pharmacokinetics of Budesonide
`(EntocortTM EC) Capsules for Crohn’s Disease. Clin Pharmacokin

This document is available on Docket Alarm but you must sign up to view it.


Or .

Accessing this document will incur an additional charge of $.

After purchase, you can access this document again without charge.

Accept $ Charge
throbber

Still Working On It

This document is taking longer than usual to download. This can happen if we need to contact the court directly to obtain the document and their servers are running slowly.

Give it another minute or two to complete, and then try the refresh button.

throbber

A few More Minutes ... Still Working

It can take up to 5 minutes for us to download a document if the court servers are running slowly.

Thank you for your continued patience.

This document could not be displayed.

We could not find this document within its docket. Please go back to the docket page and check the link. If that does not work, go back to the docket and refresh it to pull the newest information.

Your account does not support viewing this document.

You need a Paid Account to view this document. Click here to change your account type.

Your account does not support viewing this document.

Set your membership status to view this document.

With a Docket Alarm membership, you'll get a whole lot more, including:

  • Up-to-date information for this case.
  • Email alerts whenever there is an update.
  • Full text search for other cases.
  • Get email alerts whenever a new case matches your search.

Become a Member

One Moment Please

The filing “” is large (MB) and is being downloaded.

Please refresh this page in a few minutes to see if the filing has been downloaded. The filing will also be emailed to you when the download completes.

Your document is on its way!

If you do not receive the document in five minutes, contact support at support@docketalarm.com.

Sealed Document

We are unable to display this document, it may be under a court ordered seal.

If you have proper credentials to access the file, you may proceed directly to the court's system using your government issued username and password.


Access Government Site

We are redirecting you
to a mobile optimized page.





Document Unreadable or Corrupt

Refresh this Document
Go to the Docket

We are unable to display this document.

Refresh this Document
Go to the Docket