Development and validation of a Level A in-vitro in-vivo correlation for tofacitinib modified-release tablets using extrudable core system osmotic delivery technology

Development and validation of a Level A in-vitro in-vivo correlation for tofacitinib modified-release tablets using extrudable core system osmotic delivery technology

Journal Pre-proof Development and validation of a Level A in-vitro in-vivo correlation for tofacitinib modified-release tablets using extrudable core...

1MB Sizes 0 Downloads 25 Views

Journal Pre-proof

Development and validation of a Level A in-vitro in-vivo correlation for tofacitinib modified-release tablets using extrudable core system osmotic delivery technology Joseph Kushner IV , Manisha Lambaal , Thomas Stock , Ronnie Wang , Mary Anne Nemeth , Christine Alvey , Raymond Chen , Vincent DeMatteo , Andrew Blanchard PII: DOI: Reference:

S0928-0987(19)30473-7 https://doi.org/10.1016/j.ejps.2019.105200 PHASCI 105200

To appear in:

European Journal of Pharmaceutical Sciences

Received date: Revised date: Accepted date:

11 June 2019 3 November 2019 17 December 2019

Please cite this article as: Joseph Kushner IV , Manisha Lambaal , Thomas Stock , Ronnie Wang , Mary Anne Nemeth , Christine Alvey , Raymond Chen , Vincent DeMatteo , Andrew Blanchard , Development and validation of a Level A in-vitro in-vivo correlation for tofacitinib modified-release tablets using extrudable core system osmotic delivery technology, European Journal of Pharmaceutical Sciences (2019), doi: https://doi.org/10.1016/j.ejps.2019.105200

This is a PDF file of an article that has undergone enhancements after acceptance, such as the addition of a cover page and metadata, and formatting for readability, but it is not yet the definitive version of record. This version will undergo additional copyediting, typesetting and review before it is published in its final form, but we are providing this version to give early visibility of the article. Please note that, during the production process, errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain. © 2019 Published by Elsevier B.V.

Development and validation of a Level A in-vitro in-vivo correlation for tofacitinib modified-release tablets using extrudable core system osmotic delivery technology Joseph Kushner IVa,*, Manisha Lambaa, Thomas Stockb, Ronnie Wanga, Mary Anne Nemetha, Christine Alveya, Raymond Chena, Vincent DeMatteoa, Andrew Blancharda a

Pfizer Inc, 558 Eastern Point Rd, Groton, CT, 06340, USA; bPfizer Inc, 500 Arcola Road,

Collegeville, PA, 19426, USA *Corresponding author: Joseph Kushner, IV, 558 Eastern Point Rd, Groton, Connecticut 06340, USA. Telephone: +1-860-686-1098. Email addresses: [email protected] (J. Kushner), [email protected] (M Lamba), [email protected] (T. Stock), [email protected] (R. Wang), [email protected] (M. A. Nemeth), [email protected] (C. Alvey), [email protected] (R. Chen), [email protected] (V. DeMatteo), [email protected] (A. Blanchard)

1

ABSTRACT Purpose To determine if a validated Level A in-vitro in-vivo correlation (IVIVC) could be achieved with the extrudable core system (ECS) osmotic tablet platform. Tofacitinib is an oral JAK inhibitor for the treatment of rheumatoid arthritis. Methods Fast-, medium-, and slow-release modified-release formulations of 11 mg tofacitinib ECS tablets, and one formulation of 22 mg tofacitinib ECS tablet, were manufactured. In vitro dissolution of the tofacitinib ECS tablets was performed using USP Apparatus 2 (paddles) and in vivo pharmacokinetic (PK) data were obtained from a Phase 1 study in healthy volunteers. A 5 mg immediate-release formulation tablet was included to support deconvolution of the tofacitinib ECS PK tablet data to obtain the in vivo absorption profiles. A linear, piecewise correlation and a simple linear correlation were used to build and validate two IVIVC models. Results The prediction errors (PEs) for the linear, piecewise correlation met the Food and Drug Administration‟s criteria for establishing a Level A IVIVC, with a maximum absolute individual internal PE of 4.6%, a maximum absolute average internal PE of 3.9%, and a maximum absolute external PE of 8.4% obtained. Conclusions This study demonstrates that the tofacitinib ECS osmotic tablet platform can achieve a Level A IVIVC, similar to other osmotic delivery systems. KEYWORDS (6 maximum): tofacitinib · modified-release · in-vitro in-vivo correlation · extrudable core system · osmotic delivery

2

ABBREVIATIONS BCS, biopharmaceutical classification system ECS, extrudable core system IR, immediate-release IVIVC, in-vitro in-vivo correlation IVIVR, in-vitro in-vivo relation MDT, mean dissolution time MR, modified-release OROS, Oral Release Osmotic System RBA, relative bioavailability RPM, revolutions per minute SUPAC, Scale-Up and Post-Approval Changes UIR, unit impulse response USP, United States Pharmacopeia

3

1. Introduction Modified-release dosage forms are typically utilized in the pharmaceutical field to reduce the dosing frequency and/or reduce fluctuations in the plasma drug levels for therapeutic agents. Osmotic dosage forms are one class of modified-release dosage forms designed for oral delivery. Commercially-precedented types include elementary osmotic pumps, push-pull osmotic pumps, controlled porosity osmotic pumps, push-stick osmotic pumps, and drug over-coated elementary osmotic pumps (Malaterre et al., 2009). Other types of oral osmotic dosage forms include liquidOral Release Osmotic System (OROS), OROS-CT, single composition osmotic tablets, Portab, ENSOTROL®, Zero-Os, effervescent/floating osmotic pumps, sandwiched osmotic tablets, bursting osmotic tablets, push-pull osmotic capsules, asymmetric membrane tablets, and capsules (Verma et al., 2002). Dosage forms utilizing osmotic delivery technology have previously been demonstrated to have drug release profiles that are independent of the pH and the agitation rate of the surrounding media, as well as being independent of physiological factors such as food intake and patient age (Malaterre et al., 2009; Verma et al., 2002). It can be beneficial if in vitro drug release profiles are reflective of in vivo release, and an invitro in-vivo relation or correlation (IVIVR or IVIVC; defined by the Food and Drug Administration [FDA] as „a predictive mathematical model describing the relationship between an in vitro property of an oral dosage form and relevant in vivo response‟ (Kaur et al., 2015)) is commonly used for this purpose. The features of osmotic dosage forms described above make them particularly good candidates for the development of an IVIVR or IVIVC. In a systematic review conducted in 2009, 14 compounds, across a range of biopharmaceutical classification system (BCS) classifications and osmotic tablet delivery systems, were reported in the literature

4

to have demonstrated a Level A IVIVC (i.e. the highest level of correlation) in either human or animal populations (Malaterre et al., 2009). The development of an IVIVC has several benefits. During the early development of a modified-release drug product, an IVIVC provides an efficient methodology to determine the target release profile for the drug product under development, thereby streamlining future development activities. Also, a validated IVIVC can be used to support the setting of clinically meaningful dissolution ranges, via the associated predictive dissolution method, to ensure patients receive a drug product with performance similar to the drug product evaluated in the pivotal clinical and/or pharmacokinetic (PK) studies. One type of elementary or single-layer osmotic pump tablet is the extrudable core system (ECS) osmotic delivery tablet. The ECS osmotic tablet was previously described by Waterman et al and demonstrated the release of high doses of low solubility compounds from osmotic tablets (Waterman et al., 2009). Like other osmotic tablets, the ECS tablet consists of an osmoticallyformulated core containing the active agent surrounded by a semi-permeable membrane containing a delivery port through the membrane. The ECS core provides the osmotic driving force through use of a sugar or other osmotically active agent, and suspends the drug for release from the dosage form through use of high molecular weight hydroxyethyl cellulose. The release of drug from the ECS tablet was found to be most rapid when a modified oval tablet shape was used with the extrusion port included at one end of the tablet band. The performance of the ECS tablet was previously evaluated for three model weakly basic compounds and demonstrated the ability to release the active ingredient at drug loadings of up to 500 mg. An initial evaluation on the ability of the ECS tablets to provide a strong IVIVC, similar to other osmotic tablet formulations, was performed by comparing in vitro dissolution to the amount of drug released in 5

dogs. However, to date, there has been no prior demonstration of an IVIVC in humans for the ECS osmotic tablets. Tofacitinib citrate was selected as a model pharmaceutical compound to evaluate the IVIVC capability of ECS osmotic tablets. Tofacitinib is a Janus kinase inhibitor for the treatment of rheumatoid arthritis, psoriatic arthritis, and ulcerative colitis. It is currently administered twice daily (BID) as an immediate-release (IR) tablet at a dose strength of 5 mg, or once daily (QD) as a modified-release (MR) tablet at a dose strength of 11 mg, for treatment of adult patients with moderately to severely active rheumatoid arthritis and active psoriatic arthritis. Tofacitinib is also available as an IR tablet at a dose strength of 5 and 10 mg for the treatment of adult patients with moderately to severely active ulcerative colitis. To facilitate a QD posology, an ECS MR formulation has been developed. The 11 mg dose strength of the ECS MR formulation has demonstrated equivalence on total and peak exposure compared with the 5 mg IR formulation dosed BID (Lamba et al., 2016). Tofacitinib, a weakly basic compound, is classified as a BCS Class 3 compound (high solubility, moderate permeability) and has demonstrated dose proportionality for AUCinf over a range of 1–100 mg (FDA Center for Drug Evaluation and Research, 2011). The objective of the present study is to evaluate the capability of the ECS osmotic tablet to achieve a validated, Level A IVIVC in humans, using a weak base drug, tofacitinib citrate, as a model compound.

6

2. Materials and methods 2.1 Materials Tofacitinib citrate was obtained from Pfizer (Ringaskiddy, Ireland and Sandwich, UK). Sorbitol (grade Neosorb P110) was obtained from Roquette (Gurnee, Illinois). Hydroxyethyl cellulose (grade Natrosol 250 HX) was obtained from Ashland (Zwijndrecht, Netherlands). Copovidone (grade Kollidon VA 64 Fine) was obtained from BASF (Ludwigshaven, Germany). Colloidal silicon dioxide (grade Aerosil 200) was obtained from Evonik (Rheinfelden, Germany). Magnesium stearate was obtained from Mallinckrodt (St. Louis, Missouri). Cellulose acetate (grade 398-10) was obtained from Eastman (Kingsport, Tennessee). Hydroxypropyl cellulose (grade Klucel EF) was obtained from Ashland (Hopewell, VA). Acetone was obtained from Merck KGaA (grade Emprove from Darmstadt, Germany) and from Dow Chemicals (Freeport, Texas). Methanol was obtained from Pharmco (Brookfield, Connecticut). Opadry pink (grade 03K140024) was obtained from Colorcon (Humacao, Puerto Rico and Dartford, UK). Tofacitinib 5 mg IR tablets were obtained from Pfizer Inc (Freiburg, Germany). 2.2 Preparation of tofacitinib modified-release tablets Three release rates (fast, medium, and slow) of tofacitinib MR 11 mg tablets and a single release rate of tofacitinib MR 22 mg tablets (intended as a QD equivalent of IR 10 mg BID) were developed for the IVIVC study and model development. The tofacitinib MR tablets were manufactured using conventional processes for osmotic delivery tablets (Malaterre et al., 2009; Verma et al., 2002; Waterman et al., 2009), as shown in Fig. 1. The formulations are provided in Table 1. Additional manufacturing details are provided below.

7

2.2.1. Tablet core manufacture Tofacitinib MR 11 mg tablet cores were manufactured using a direct compression process. Tofacitinib citrate, sorbitol, hydroxyethyl cellulose, and copovidone were mixed using a blendmill-blend process. An 800 liter bin blender was operated for 20 minutes at 12 rpm for both blending steps. After the first blending step, the blend was passed through a CoMil (Model 197S) operating at 1,400 rpm. After the second blend step, magnesium stearate was added to the top of the blend and mixed into the blend for 5 minutes at 12 rpm. The blend was compressed into modified oval tablets with a target weight of 200 mg using a Manesty Mark IV rotary tablet press. Tofacitinib MR 22 mg tablet cores were also manufactured using a direct compression process. Tofacitinib citrate, sorbitol, hydroxyethyl cellulose, copovidone, and colloidal silicon dioxide were mixed using a blend-mill-blend process. A 28 liter bin blender was operated for 20 minutes at 12 rpm for both blending steps. After the first blending step, the blend was passed through a Bohle Turbo Mill (Model BTS200) operating at 1,500 rpm. After the second blend step, magnesium stearate was added to the top of the blend and mixed into the blend for 5.25 minutes at 12 rpm. The blend was compressed into modified oval tablets with a target weight of 200 mg using a Fette PT1200 rotary tablet press. 2.2.2. Application of functional coating The functional coating solution was prepared by mixing acetone, methanol, cellulose acetate, and hydroxypropyl cellulose together. A 250 kg batch of tofacitinib MR 11 mg medium-release tablets were spray coated with a Vector Hi-Coater, using conventional spray coating methods. 900 gram batches of tofacitinib MR 11 mg fast-release, slow-release, and MR 22 mg tablets were 8

spray coated with Vector LCDS, using conventional spray coating methods. The different tablet release rates for the MR 11 mg were obtained by varying the amount of the rate-controlling, semi-permeable coating applied to the tablet cores using the different scales of coating equipment. 2.2.3. Laser drilling and drying A 600 micron extrusion port was laser-drilled into the tofacitinib MR 11 mg medium-release tablets using a rotary laser drill (EAM Corp, Brooklyn, NY). The extrusion port was located on the end of the tablet band, as described in Waterman et al (Waterman et al., 2009). These tablets were then dried for 24 hours at 40oC to minimize residual solvents. For the tofacitinib MR 11 mg fast-release, slow-release, and MR 22 mg tablets, the tablets were first dried for 16 hours at 40oC. A 600 micron extrusion port was laser-drilled into these dried tablets using an X-Y station laser drill (Bend Research, Bend, OR). The extrusion port was located on the end of the tablet band, as described in Waterman et al (Waterman et al., 2009). 2.2.4. Aqueous color coating The color coating suspension was prepared by mixing Opadry pink with water. A 120 kg batch of drilled and dried tofacitinib MR 11 mg medium-release tablets were coated with the color coating suspension using an AC48 tablet spray coater, using conventional spray coating methods. 200–300 gram batches of the drilled and dried tofacitinib MR 11 mg fast- and slow-release tablets, and MR 22 mg tablets were coated with the color coating suspension using a Vector LDCS tablet spray coater, using conventional spray coating methods. Sufficient coating was applied to ensure that there was a uniform color on the surface of the tablets.

9

2.3. In vitro dissolution testing In vitro dissolution testing for the MR tablets was performed using United States Pharmacopeia (USP) Apparatus 2 (paddles) at 50 rpm in 0.9 liters of pH 6.8, 50 mM phosphate buffer at 37oC with sinkers. The dissolution test condition was selected based on results from experimental DoE studies, which examined the impact of the Apparatus type (Type 1 baskets or Type 2 paddles), stirring speed (50, 75, and 100 rpm), and pH. Apparatus 2 (paddles) was selected over Apparatus 1 (baskets) to minimize variability. The paddle speed of 50 rpm was selected as the most conservative agitation option. The buffer of 50 mM potassium phosphate at pH 6.8 was chosen as the in vivo drug release is expected to occur in the small intestinal region, where the pH is approximately neutral. The amount of tofacitinib dissolved was assayed using an in situ ultraviolet fiber-optic system (OPT-DISS UV FiberOptic, LEAP Technologies, Carrboro, NC) with 10 mm Arch probes. Twelve units were tested from each of the modified-release formulations described previously. 2.4. In vivo pharmacokinetics study In vivo pharmacokinetics (PK) data were obtained from a Phase 1, randomized, open-label, 5treatment, 5-period, 6-sequence partial crossover study conducted in 36 healthy volunteers (Pfizer study A3921195). The first three periods consisted of a full 3-way crossover of the three tofacitinib MR 11 mg formulations (fast-, medium-, and slow-release rates) to ensure that the three formulations intended for internal validation of the IVIVC model were protected from potential subject drop-outs during the last two periods of the study. Each of the first three periods was followed by a washout period of ≥72 hours. A 5 mg dose of the IR tablet formulation of tofacitinib was included in the fourth period to serve as the unit impulse response (UIR) to

10

enable deconvolution of individual-level PK data from the modified-release formulations. The IR tablet formulation was selected as the UIR due to the high solubility and rapid dissolution of tofacitinib from this dosage form. After a wash-out period of ≥24 hours following dosing in the fourth period, a 22 mg dose of the MR formulation of tofacitinib was administered in the fifth period to support external validation of the Level A IVIVC. Serial plasma samples, for determination of tofacitinib PK parameters, were collected pre-dose and up to 72 hours post-dose following each MR treatment and up to 24 hours post-dose following the IR treatment, as follows: PK sampling for the MR treatments was 0.5, 1, 2, 3, 4, 6, 8, 12, 24, 36, 48, and 72 hours post-dose, and for the IR treatment: 0.5, 1, 2, 3, 4, 6, 8, 12, and 24 hours post-dose. Since tofacitinib has demonstrated dose proportionality over the range of 1–100 mg (FDA Center for Drug Evaluation and Research, 2011), the use of three different dose strengths (i.e. 5, 11, and 22 mg) is not expected to have a significant impact on the interpretation of the study results. This study was conducted in accordance with the Declaration of Helsinki and the International Conference on Harmonization Good Clinical Practice Guidelines, and was approved by the Institutional Review Boards and/or Independent Ethics Committees at the investigational center. All subjects provided written informed consent. 2.5. Statistical analyses A sample size of 30 evaluable subjects (5 per sequence) for the in vivo PK study was chosen to provide 85% coverage probability that the upper bound of the 90% confidence interval (CI) for the true mean Cmax ratio of 90% for slow-release to medium-release should fall below 100%, and at least 99.9% coverage probability that the lower bound of 90% CI for the true mean C max ratio of 120% for fast-release to medium-release should exceed 100% for MR 11 mg formulations.

11

This metric would ensure the ability to adequately differentiate the in vivo performance of the various formulations. Considering possible drop-outs, a total of 36 healthy volunteers were enrolled in the study. In addition to developing an IVIVC (described in the section below), standard estimates for relative bioavailability were estimated as follows. Natural log-transformed AUCinf and Cmax from the first three periods (MR 11 mg tablets) were analyzed using a mixed-effects model with treatment, sequence, and period as fixed effects and subject within sequence as the random effect. Estimates of the adjusted mean differences (test/reference) and corresponding 90% CIs were obtained from the model. The adjusted mean differences and 90% CIs for the differences were exponentiated to provide estimates of the ratio of adjusted geometric means (test/reference) and 90% CIs for the ratios. Three relative bioavailability (RBA) assessments were conducted. In the first, comparing MR 11 mg formulations, the medium-release formulation was the reference. The slow-release and fast-release formulations were the test treatments. In the second RBA comparison, the slowrelease formulation was the reference and the fast-release formulation was the test treatment. In the third RBA assessment, dose-normalized exposures for the MR 11 mg medium-release, MR 11 mg slow-release and MR 22 mg were compared and the proportionality of exposures between the two dose strengths was evaluated using another mixed-effects model with treatment as the fixed effect and subject as the random effect.

12

2.6. Development and validation of the Level A IVIVC model 2.6.1. Modeling software Phoenix WinNonLin, Build 8.0.0.3176 (Certara, Princeton, NJ) was used to perform the development and validation of the Level A IVIVC for the tofacitinib MR tablets. 2.6.2. In vitro dissolution modeling The Weibull equation (Equation [1]) was used to model the dissolution data in WinNonLin.

(1)

Fdiss ( t )  int  (F  int)  [1  e

 (( t  t lag ) / MDT ) b

]

Where Fdiss(t) = percent of drug dissolution as function of time, F = amount released at time infinity, tlag = lag time to onset of release, in the preferred units of time, MDT = mean dissolution time, in the preferred units of time, b = slope factor (no units), and int = y-intercept, zero (default), or estimated intercept, using the preferred units for Fdiss. The asymptotic fraction dissolved, F, is fixed to 1.0 and the y-intercept, int, is fixed to 0 to prevent incomplete measurements of dissolution. Each mean in vitro dissolution profile was fitted to the Weibull formula using non-linear, least squares regression with MDT, b, and t lag as the estimated parameters. 2.6.3. Deconvolution of in vivo pharmacokinetic data The in vivo absorption profiles were obtained by a numerical deconvolution procedure on the individual tofacitinib concentration-time profile data of the MR tablets, executed within WinNonLin. The individual plasma tofacitinib concentration-time data from the immediate13

release tablet at 5 mg dose strength was used to generate the UIR from the individual subjects. A maximum of 3 UIR exponentials were allowed and a time lag was included. Deconvolution was then performed, using 289 output points which would provide an output point every 15 minutes for the 72-hour MR PK profiles. 2.6.4. Development and validation of IVIVC model The internal validation or predictability is defined as how well the proposed IVIVC model predicted the in vivo performance of each formulation used in building the model. External validation was also used to evaluate predictability for formulations not used in the development of the model. Evaluating the internal and external validation of the model was conducted under two scenarios to evaluate the robustness of the IVIVC for the tofacitinib ECS tablets. In the first scenario, all three MR 11 mg formulations (fast-, medium-, and slow-release) served as internal validation, while the MR 22 mg formulation served as external validation. In the second scenario, the fast- and slow-release MR 11 mg formulations served as internal validation, while MR 11 mg medium-release and MR 22 mg served as external validation. In both scenarios, the external validation formulations were omitted from the model building process. As such, the MR 22 mg formulation was not included for development of the IVIVC model. The predicted in vivo plasma tofacitinib concentration versus time profile for the external validation formulations was determined from the IVIVC obtained from internal validation formulations with different release rates, along with the dissolution profiles of the external validation formulations. The individual plasma tofacitinib concentration profiles from tofacitinib MR tablets with varying release rates and dose strengths were simulated by applying the dissolution data from these formulations to the IVIVC model (5-parameter user-built model) to predict the absorption

14

profiles followed by the convolution of the absorption profile with the UIR function. The userbuilt model, presented in Equation (2), is a piecewise, linear function that models the fraction of drug absorbed, Fabs, as a function of time, t, in the domain of high absorption, the domain of reduced absorption, and the domain where in vitro dissolution has been cut off.

(2)

Fdiss timescale 1 * t , t  t 1  Fabs  AbsScale * Fdiss timescale 1 * t 1  timescale 2 * t  t 1 , t 1  t  t cutoff F timescale * t  timescale * t 1 1 2 cutoff  t 1 , t  t cutoff  diss

The parameters in this model include: 

AbsScale: absorption scaling factor



t1: time (in hours) at which drug absorption in vivo transfer from the region of high absorption to the region of reduced absorption



timescale1: in vitro time scaling factor for the region of high absorption



timescale2: in vitro time scaling factor for the region of reduced absorption



tcutoff: time (in hours) after which dissolution (“Diss”) should stop increasing.

This modeling approach is similar to the piecewise, linear correlation models recently proposed by Kakhi et al for formulations whose release rate was controlled by the thickness of a semipermeable coating (Kakhi et al., 2013) and by Kesisoglou et al for matrix and multiparticulate formulations (Kesisoglou et al., 2015). The primary difference in the present model is the use of two time-scaling factors, rather than two absorption-scaling factors used by the previous authors.

15

This step-wise model incorporating the potential for different regions of absorption was utilized based on results of an earlier study (A3921113), which showed the relative bioavailability of tofacitinib modified-release formulations decreased as the duration of drug release was prolonged (FDA Center for Drug Evaluation and Research, 2015). A simple linear model (i.e. Fabs = AbsScale * Fdiss [timescale * t]) with a single absorption scaling factor and a single time scaling factor was also evaluated as a comparison with the piecewise linear model. Convolution of the absorption profiles was performed with 145 output points to provide a predicted output point at each hour of the predicted PK profile to facilitate a more direct comparison to the PK sampling time points used in the Phase 1 in vivo study (A3921195). AUCinf and Cmax values were determined from these simulated PK profiles by use of standard noncompartmental methods. The % prediction error (%PE) defined in Equation (3) and Equation (4) were calculated for each formulation.

Cmax (obs)  Cmax (pred)  100% Cmax (obs)

(3)

%PECmax 

(4)

%PE AUCinf 

AUCinf (obs)  AUCinf (pred)  100% AUCinf (obs)

Where Cmax (obs) and Cmax (pred) = the geometric mean observed and IVIVC model predicted maximum plasma concentration, respectively; and AUCinf (obs) and AUCinf (pred) = the geometric mean observed and IVIVC predicted AUCinf for the plasma concentration profiles, respectively. 16

For internal validation, the proposed IVIVC model was considered validated if the average absolute %PE across all internal validation formulations was 10% or less for both Cmax and AUCinf and if the %PE for each formulation did not exceed 15%. For external validation, the proposed IVIVC model was considered validated if the %PE of the external formulations was 10% or less for both Cmax and AUCinf. These validation criteria are consistent with the FDA guidance on IVIVCs for modified-release oral dosage forms (Food and Drug Administration, 1997a). 3. Results 3.1. In vitro dissolution Mean dissolution profiles for the fast-release, medium-release, and slow-release formulations of tofacitinib MR 11 mg, and for the MR 22 mg formulation, are shown in Fig. 2. The fast-release and slow-release tofacitinib MR 11 mg tablets showed a mean difference of 25% drug dissolved at the 2.5 hour time point (mean range: 36% to 61%, and a value of 50% for medium-release). The tofacitinib MR 22 mg tablet exhibited in vitro dissolution performance at the 2.5 hour time point within the range observed for the tofacitinib MR 11 mg tablet formulations. The similarity of the shape of the dissolution profiles indicated that tofacitinib release was comparable across all formulations and essentially controlled by water permeation through the semi-permeable tablet coating under osmotic pressure. The comparison between fitted dissolution profiles and observed dissolution results for MR 11 mg formulations and MR 22 mg formulation are presented in Fig. 3. Excellent agreement between fitted and observed values was obtained. Table 2 lists the regressed parameter values for tlag, MDT, and int, as well as the goodness of fit, coefficient of determination (R 2), for each 17

tofacitinib MR formulation. The in vitro dissolution profiles were well-modeled by the Weibull equation, as indicated by the high R2 values for each formulation. Furthermore, the three model parameters (MDT, b, and t lag) show clear differences in the mean in vitro dissolution profiles for the fast- and slow-release MR 11 mg formulations. For the MDT model parameter, there is also clear differentiation of the medium-release MR 11 mg formulation from both the fast- and slowrelease MR 11 mg formulations. 3.2. In vivo pharmacokinetics Thirty-six healthy male adults (aged 21–49 years of age) were enrolled and completed the study. Tofacitinib plasma concentration–time profiles for all treatments are shown in Fig. 4. Consistent with the design targets of different MR formulations, the fast-release MR 11 mg formulation achieved a higher peak concentration (geometric mean: 44.83 ng/ml) than both the mediumrelease (41.40 ng/ml) and the slow-release MR 11 mg formulation (37.23 ng/ml; Table 3). The fast-release formulation also achieved an earlier Tmax with a median value of 3 hours, versus 4 hours for both the medium- and slow-release MR 11 mg formulations. The mean terminal t 1/2 was 5.8 hours for the fast-release, 5.5 hours for the medium-release, and 5.9 hours for the slowrelease MR 11 mg formulations. As expected for the IR 5 mg formulation, it achieved peak concentration (geometric mean: 48.14 ng/ml) rapidly (0.5 hours post-dose); elimination t 1/2 was ~ 3 hours (Table 3). The difference in the terminal t 1/2 between the MR and IR formulations could be due to the occurrence of flip-flop kinetics, where the rate of elimination is limited by the absorption rate of the tofacitinib MR formulation. The 90% CIs for the ratios of adjusted geometric means (slow-release vs medium-release, and fast-release vs medium-release) for AUCinf were contained within 80% to 125% equivalence 18

limits (Table 4). Additionally, while the Cmax ratio for slow- and fast-release versus mediumrelease was contained within the equivalence limits, the 90% CIs excluded 100% for both formulations, demonstrating an in vivo difference in the rate of absorption between both the fastrelease and slow-release formulations relative to the medium-release MR 11 mg formulation. Further, the 90% CIs for the ratio of adjusted geometric means for fast-release versus slowrelease for AUCinf were also contained within 80% to 125% equivalence limits. For Cmax, the ratio of adjusted geometric means for fast-release versus slow-release was 120.41%, with 90% CIs of 112.88% to 128.45%. Finally, dose-normalized Cmax and AUCinf values for the MR 11 mg medium-release, MR 11 mg slow-release, and MR 22 mg tablets were similar, which is in agreement with prior results obtained in Study A3921132, demonstrating dose proportionality between the MR 11 mg and MR 22 mg dose strengths (FDA Center for Drug Evaluation and Research, 2015). The 90% CIs for the ratios of geometric means (MR 22 mg vs MR 11 mg medium-release and MR 22 mg vs MR 11 mg slow-release), for both AUCinf and Cmax, were contained within the 80% to 125% equivalence limits. 3.3. IVIVC model development and validation using the piecewise linear model The mean absorption profiles for the three MR 11 mg tablets and MR 22 mg tablets are shown in Fig. 5. The in vivo absorption profiles from deconvolution analysis trend with the rank-order of the in vitro dissolution profiles (i.e. MR 11 mg fast-release, MR 11 mg medium-release, and MR 11 mg slow-release, with MR 22 mg similar to MR 11 mg slow-release). A Levy plot, presented in Fig. 6, was generated by using Phoenix WinNonlin to compare in vivo absorption versus in vitro dissolution data visually, as time-versus-time plots matched values for absorption and dissolution. The plot includes a linear fit of the data points with intersection at origin, slope of 0.7945, and correlation line (R2 = 0.9966). 19

The regressed values for each of the five parameters in the user-built piecewise linear model from the correlation development are listed in Table 5a. In the case where all three MR 11 mg formulations served as the internal validation treatments, the correlation between fraction dissolved and fraction absorbed with parameters listed provides an R 2 value for the MR 11 medium-, fast-, and slow-release tablet data of 99.76%, 99.83% and 99.78%, respectively. In the case where the fast- and slow-release MR 11 mg formulations serve as the internal validation treatments, the correlation between fraction dissolved and fraction absorbed with parameters listed provides an R2 value for the MR 11 mg fast- and slow-release tablet data of 99.82% and 99.86%, respectively. Validation results for the piecewise linear model are presented in Tables 6a and 7a. For the case of all three MR 11 mg formulations being used for internal validation (Table 6a), %PEs for both AUCinf and Cmax for internal validation formulations were less than or equal to ±2.4% for all the observed values of the three release rates, and less than or equal to ±1.9% on average. For external validation, %PEs for AUCinf and Cmax values for tofacitinib MR 22 mg tablets were less than or equal to ±6.2%, also reported in Table 6a. For the case of only the fast- and slow-release MR 11 mg formulations used for internal validation, %PEs for AUCinf and Cmax for internal validation (Table 7a), were less than or equal to ±4.6% for all the observed values of the two release rates, and less than or equal to ±3.9% on average. For external validation, %PEs for AUCinf and Cmax values for tofacitinib MR 11 mg medium-release and MR 22 mg tablets were less than or equal to ±8.4%, also reported in Table 7a. Overall, using either modeling approach, the obtained Level A IVIVC model predicted AUCinf and Cmax to be well within ±10% of the observed values for both internal and external validation. The quality of the IVIVC piecewise

20

linear models is further confirmed in Fig. 7, which compares the IVIVC model-predicted PK profiles to the observed PK profiles obtained from the clinical study. 3.4 IVIVC model development and validation using the simple linear model Table 5b presents the correlation parameters for the two cases where a simple linear model was used to correlate the absorption and dissolution profile data. The correlation between fraction dissolved and fraction absorbed with the parameters listed in Table 5b provided an R2 value for the MR 11 mg medium-, fast-, and slow-release tablet data of 98.40%, 99.32%, and 99.01%, respectively, for the case when MR 11 mg medium-release is an internal validation treatment, and an R2 value for the MR 11 mg fast- and slow-release tablet data of 99.30% and 99.01%, respectively, for the case when MR 11 mg medium-release is an external validation treatment. Tables 6b and 7b present the validation results for the simple linear model. When MR 11 mg medium-release was included in the internal validation (Table 6b), the simple linear model met the internal validation criteria, but the MR 22 mg formulation for external validation had a %PE value for Cmax higher than the threshold of ±10% (i.e. -11.7%), which did not meet the external validation criteria. When MR 11 mg medium-release was considered as another external validation treatment (Table 7b), the internal validation of the simple linear model was also able to meet the internal validation criteria, but the MR 22 mg formulation for external validation also had a %PE value for Cmax higher than the threshold of ±10% (i.e. -12.9%), which did not meet the external validation criteria. 4. Discussion A typical prerequisite for developing an IVIVC is the development of formulations with sufficient difference in the rates of release to achieve observation of difference in the PK 21

performance of the evaluated formulations (Food and Drug Administration, 1997a). This prerequisite was achieved for the three MR 11 mg formulations based on the differences in the % dissolved at 2.5 hours (i.e. greater than 10% difference) and the differences in the Weibull model parameters for these three formulations. Differentiated release rates can be achieved by modulation of the excipient(s) that impart the release rate controlling mechanism to the modified-release formulation. For osmotic delivery tablets, the release rate of the drug is controlled by the rate of water permeation through the semi-permeable membrane applied to the osmotically active tablet core and can be described by the following equation (Verma et al., 2002):

(5)

dM A  L p   p C dt h

where dM/dt is the rate of drug release from the formulation, A and h are the surface area and thickness of the semi-permeable membrane, Lp is the water permeability of the semi-permeable membrane,  is the reflection coefficient,  is the osmotic pressure difference, p is the hydrostatic pressure difference, and C is the concentration of the drug in the formulation. Equation (5) can be further simplified under the following assumptions: 1)  is approximately unity for perfect semi-permeable membranes, which only allow passage of water and prevent passage of other solutes, 2)  may be approximated for , the osmotic pressure of the osmotic agent within the tablet core, since this value is typically greater than the osmotic pressure of the surrounding in vitro or in vivo media, and 3) p is small compared to osmotic pressure for a formulation with a sufficiently large delivery orifice (2). With these assumptions, Equation (5) can be simplified as follows:

22

(6)

dM A  L pC dt h

For the tofacitinib MR formulations evaluated in the present study, the drug (tofacitinib), osmogen (sorbitol), and size and shape of the tablets were the same across all formulations. Therefore, the values for A, C, and  in Equation (6) are the same across all of the MR 11 mg formulations. For the MR 22 mg formulation, A and  are also the same as the MR 11 mg formulations, while C is doubled. However, this factor of 2 is addressed by reporting the in vitro dissolution data for all tofacitinib MR formulations in terms of the percentage of tofacitinib dissolved, which normalizes the tofacitinib MR 22 mg and 11 mg dissolution data. This implies that only the membrane thickness and the water permeability were modulated to provide the different release rates of tofacitinib from these MR formulations. The water permeability and thickness of this membrane can be modulated by both formulation (e.g. composition of the coating, amount of coating applied) and process factors (e.g. scale of the functional coating process). In the present study, differences in the amount of coating applied and the scale of the functional coating process (lab-scale vs commercial-scale) were modulated to achieve the in vitro dissolution release profiles provided in Fig. 2, while the composition (e.g. the ratio and grades of cellulose acetate to hydroxypropyl cellulose) was held constant. For the lab-scale functional coating of the MR 11 mg tablets, the fast- and slow-release formulations were obtained by modulating the amount of functional coating applied. Specifically, 10.4 mg and 15.2 mg of coating were applied to obtain the fast- and slow-release formulations, respectively, which translate to a modulation of the membrane thickness. For the MR 11 mg medium-release tablets, 23

the functional coating was applied using a commercial-scale tablet coater. To achieve the medium-release profile, the amount of coating required with the commercial-scale equipment was 16.4 mg. This amount of coating is greater than the amount of coating applied to achieve the slow-release formulation with lab-scale coating equipment, and further suggests that the medium-release tablet has a thicker semi-permeable membrane compared to the lab-scale slowrelease formulation. According to Equation (6), to achieve a faster release rate than the slowrelease formulation, a more permeable functional coating (i.e. larger value of L p) was applied to the medium-release formulation with the selected commercial-scale equipment than was applied to the slow-release tablets with the selected lab-scale coating equipment. Permeability differences resulting from modulation of the micro-structure of the functional coating (e.g. changes in the porosity and tortuosity of the water diffusion pathways in the semi-permeable membrane) can occur across different scales of tablet pan coating equipment as the dynamics of spray droplet application to the tablet surface and the subsequent drying of the droplet on the tablet surface may differ across the scales of manufacture, due to their dependence on the processing conditions (e.g. pan speed, spray rate, atomization pressure, dew point, inlet air temperature, etc. (am Ende et al., 2000; am Ende and Berchielli, 2005)). Based on the results for the present formulations, the lower membrane permeability obtained with the lab-scale coating equipment suggests a membrane structure with reduced membrane porosity, increased tortuosity of the water diffusion path length, or both, which would reduce the effective diffusion coefficient of water through the membrane. While models and experimental approaches are available to improve the similarity of membrane properties across different manufacturing scales (Pandey and Bindra, 2014; Pandey et al., 2006; Shamblin, 2010), this aspect presents a significant technical challenge and was deemed outside of the scope of the present study.

24

For the development of an IVIVC model for a BCS Class 3 drug, Kesisoglou et al (Kesisoglou et al., 2015) found that a piecewise linear correlation provided improved %PE values compared to a traditional time scale/shift model. A similar outcome was observed in the present study, where the simple linear model was unable to meet the external validation criteria to achieve Level A validation. The motivation for utilizing a piecewise linear correlation was the observation of reduced bioavailability with the slowest release formulation, which was proposed to result from regional absorption of the BCS Class 3 compound evaluated in their study (Kesisoglou et al., 2015). Use of the piecewise linear correlation enabled the ability to incorporate two different absorption domains into the correlation, reflecting the relatively high absorption domain in the upper GI and the relatively lower absorption domain in the lower GI (Kesisoglou et al., 2015). Kesisoglou et al captured the difference in the absorption domains of the GI by introducing two absorption scaling factors, one for the domain of high absorption and one for the domain of low absorption (Kesisoglou et al., 2015), similar to an approach first proposed by Kakhi et al (Kakhi et al., 2013). Since the model compound evaluated in the present study, tofacitinib, is also classified as a BCS Class 3 drug and previously showed a decrease in AUCinf as the duration of release from the controlled release formulations increased (FDA Center for Drug Evaluation and Research, 2015), a piecewise linear correlation was selected for development of the IVIVC. However, the present modeling approach differentiated the two domains of absorption in the piecewise linear correlation through the use of two time scaling factors, rather than two absorption scaling factors. Functionally, the use of two time scaling factors achieves the same effect (e.g. differentiating the domain of high absorption from the domain of low absorption), as using two absorption scaling factors. The time at which the transition from the domain of high absorption to the domain of 25

low absorption occurred in the Kesisoglou et al analysis (i.e. 5.12 hours for matrix tablets and 5.37 hours for multiparticulates) (Kesisoglou et al., 2015) is similar to the transition time obtained from the present modeling approach using two time scaling factors (i.e. 5.20 hours for three MR 11 mg formulations for internal validation and 5.63 hours for only fast- and slowrelease MR 11 mg formulations for internal validation). From a modeling standpoint, the variable absorption factor used by Kesisoglou et al (Kesisoglou et al., 2015) and Kakhi et al (Kakhi et al., 2013) assumes that there is no change in the rate of drug dissolution as the modified-release dosage form transits through the GI tract. For osmotic tablets, release of the drug from the dosage form is generally considered to be independent of pH effects and mixing dynamics (Verma et al., 2002). However, if the drug is released from the tablet in an undissolved form, dissolution of the drug must still occur prior to absorption. Since fluid content in the large intestine was found to be reduced compared to the small intestine fluid content (Schiller et al., 2005), a reduced rate of drug dissolution could also contribute to reduced drug absorption in the lower GI, in addition to the reduced surface area available for absorption. Therefore, the use of variable time scaling factors also allows for potential impacts of drug dissolution rate to be considered. The robustness of the IVIVC for the ECS osmotic tablet platform containing tofacitinib was further demonstrated by meeting the criteria for a validated Level A IVIVC with two combinations of internal and external treatments in the IVIVC model development and validation steps when the piecewise linear model was utilized. Minimal differences were observed in the values of the correlation model parameters reported in Table 5. The difference in the parameter values for timescale1 were within the 95% CI, and, therefore, are not significantly different. For the other four model parameters (AbsScale, t1, timescale1, and timescale2), the differences 26

between the values using the two combinations was within 8%, with the greatest difference observed in the t1 parameter for the time to transition from high to low absorption regions. These minimal differences translated to minor differences (i.e. 2.2% difference or less) in the %PE values for each formulation between the two approaches reported in Tables 6 and 7. The slight increases observed in average absolute internal %PE values for Cmax and AUCinf for the second combination, where MR 11 mg fast and slow-release are the only formulations used for internal validation, are possibly a result of only two formulations being included in the model correlation building. While developing a validated IVIVC with only these two formulations is the most challenging case, inclusion of the third release rate improves the ability of the model to predict the PK performance from the dissolution data, by reducing the leverage that the two extreme formulations have on the correlation and the corresponding model parameters. Further, the use of the two time scaling factors in the piecewise linear correlation for the tofacitinib IVIVC was able to meet both the internal and external validation criteria. This degree of validation was not previously demonstrated by the variable absorption factor approach (Kakhi et al., 2013; Kesisoglou et al., 2015). Inconclusive external validation of the medium-release formulation was observed when only fast and slow formulations were included in the IVIVC model development reported by Kakhi et al (Kakhi et al., 2013). In the Kesisoglou et al study, internal validation criteria were met only for the multiparticulate formulations, using an IVIVC model generated from the multiparticulate formulations (Kesisoglou et al., 2015). This multiparticulate model was unable to meet external validation criteria when tested against the fast- and slow-release matrix formulations (Kesisoglou et al., 2015). No external validation of the Kesisoglou multiparticulate model was performed against additional multiparticulate formulations (Kesisoglou et al., 2015). In this study, a similar inconclusive external validation result was obtained when the simple

27

linear model was used to build the model correlation, as shown in Tables 6 and 7 where the MR 22 mg Cmax %PE exceeded the threshold of ±10%. Having demonstrated that a Level A IVIVC can be achieved with the ECS osmotic tablet technology, it then follows that a drug product utilizing ECS osmotic tablet technology may also benefit from utilizing a Level A IVIVC to support biorelevant dissolution specification setting and biowaivers of future bioequivalence studies to support post-approval changes, similar to other modified-release dosage forms. In the present study, three release rates of the MR 11 mg ECS osmotic tablets were used to develop the Level A IVIVC. This range of release rates had the greatest impact on Cmax (20% change, see Table 3), while the impact on AUC was much less (5% change, see Table 3). These results suggest that, for the present case, C max would be the limiting PK parameter for dissolution specification setting to ensure bioequivalence across the proposed range of dissolution specifications. Currently, FDA guidance suggests that biorelevant dissolution specifications should be set to ensure no more than a 20% difference in either C max or AUC between upper and lower specification limits (Food and Drug Administration, 1997a). However, this criterion only considers the difference in mean Cmax and AUC values, while the criterion for average bioequivalence requires that the 90% CIs for the ratio of test and reference treatments are wholly within the interval of 80–125% (Food and Drug Administration, 2001). Recently, Roudier et al proposed an approach for setting dissolution criteria with consideration for the variability of the compound being evaluated (Roudier et al., 2014). Such an approach may enable the setting of broader dissolution specifications beyond the criteria that there is no more than a 20% difference in either Cmax or AUC, while maintaining confidence in the bioequivalent performance across the range of dissolution profiles allowed by the specifications.

28

Based on FDA guidance documents, a Level A IVIVC enables biowaivers for Scale-up and PostApproval Changes (SUPAC) to modified-release dosage forms, which would normally require a bioequivalence study to support such Level 3 manufacturing changes (Food and Drug Administration, 1997a, b). These changes may include: Level 3 changes in the amounts of the non-release rate controlling excipients; Level 3 changes in the amounts of the release rate controlling excipients; Level 3 change in the site of manufacture; and Level 3 change in the manufacturing process (Food and Drug Administration, 1997a, b). Further, when an IVIVC is validated, it can also be used to reduce the amount of dissolution testing required to support Level 2 SUPAC modifications. In the absence of a validated IVIVC, dissolution data with the compendial method and in three other media are required to support the proposed modification. However, with a validated IVIVC, dissolution data is only required with the correlating in vitro dissolution method. Therefore, a validated IVIVC has value as a rational and efficient means to satisfy regulatory considerations for SUPAC modifications to dosage forms. 5. Conclusions This was the first demonstration of a validated Level A IVIVC in humans for the ECS osmotic delivery technology. Further, the results indicate the tofacitinib modified-release tablets using ECS osmotic delivery technology meet the requirements for a validated Level A IVIVC with the user-built piecewise linear model, whereas the simple linear model was not able to meet the Level A IVIVC external validation criteria. The evaluated in vitro dissolution method can serve as a predictor of in vivo performance, thereby enabling identification of clinically relevant dissolution acceptance criteria and pursuit of biowaivers for potential changes in the manufacture of the tofacitinib ECS drug product.

29

Acknowledgments The authors would like to thank the following people for their support: Alfred Berchielli, John Heimlich, Ruchi Thombre, Scott Herbig, Mahesh Krishnan, John Larmann, Loren Wrisley, Ling Zhang, Jose Alvarez, Aisdaly Martinez, Mariely Martinez, Michelle Collins, Mark Culver, and the members of the Pfizer Sandwich Solid Dosage Manufacturing and Barceloneta Technical Service and Manufacturing groups for their support in preparing the clinical study supplies; Ravi Shanker, Lisa Yuhas, Rong Li, and Jack Cook for technical guidance and support with IVIVC model development; and the colleagues of Pfizer Brussels Clinical Research Unit who conducted the study. Competing interests Christine Alvey, Thomas Stock, and Manisha Lamba were employees of Pfizer Inc at the time the study was conducted. The remaining authors are current employees and shareholders of Pfizer Inc. Data sharing statement Upon request, and subject to certain criteria, conditions, and exceptions see (https://www.pfizer.com/science/clinical-trials/trial-data-and-results for more information), Pfizer will provide access to individual de-identified participant data from Pfizer-sponsored global interventional clinical studies conducted for medicines, vaccines, and medical devices (1) for indications that have been approved in the US and/or EU or (2) in programs that have been terminated (i.e., development for all indications has been discontinued). Pfizer will also consider requests for the protocol, data dictionary, and statistical analysis plan. Data may be requested

30

from Pfizer trials 24 months after study completion. The de-identified participant data will be made available to researchers whose proposals meet the research criteria and other conditions, and for which an exception does not apply, via a secure portal. To gain access, data requestors must enter into a data access agreement with Pfizer. Funding This work was supported by Pfizer Inc. Medical writing support, under the direction of the authors, was provided by Kirsten Woollcott, MSc, at CMC Connect, a division of McCann Health Communications Ltd., Glasgow, UK, and Carole Evans, PhD, on behalf of CMC Connect, and was funded by Pfizer Inc, New York, NY, USA in accordance with Good Publication Practice (GPP3) guidelines (Ann Intern Med 2015; 163: 461–464).

31

References

am Ende, M., Herbig, S., Korsmeyer, R., 2000. Osmotic drug delivery from asymmetric membrane film-coated dosage forms, in: Wise, D.L. (Ed.), Handbook of Pharmaceutical Controlled Release Technology. CRC Press, Oxford, pp. 751-785. am Ende, M.T., Berchielli, A., 2005. A thermodynamic model for organic and aqueous tablet film coating. Pharm. Dev. Technol 10, 47-58. FDA Center for Drug Evaluation and Research, 2011. Clinical pharmacology and biopharmaceutics review of tofacitinib. NDA203214. Available at: http://www.accessdata.fda.gov/drugsatfda_docs/nda/2012/203214Orig1s000ClinPharmR.pdf. Accessed 6/14/2016. FDA Center for Drug Evaluation and Research, 2015. Clinical pharmacology and biopharmaceutics review of tofacitinib. NDA208246. Available at: https://www.accessdata.fda.gov/drugsatfda_docs/nda/2016/208246Orig1s000ClinpharmR.pdf. Accessed 3/21/2017. Food and Drug Administration, 1997a. Guidance for Industry: Extended release oral dosage forms: development, evaluation and application of in vitro/in vivo correlations. U.S. Department of Health and Human Services Food and Drug Administration Center for Drug Evaluation and Research, pp 1-24. Food and Drug Administration, 1997b. Guidance for Industry: SUPAC-MR: modified release solid oral dosage forms. Available at: http://www.fda.gov/downloads/Drugs/Guidances/UCM070640.pdf. Accessed 3/21/2017. 32

Food and Drug Administration, 2001. Guidance for Industry: Statistical approaches to establishing bioequivalence. US Department of Health and Human Services, Food and Drug Administration Center for Drug Evaluation and Research, pp 1-45. Kakhi, M., Marroum, P., Chittenden, J., 2013. Analysis of level A in vitro-in vivo correlations for an extended-release formulation with limited bioavailability. Biopharm. Drug Dispos 34, 262-277. Kaur, P., Jiang, X., Duan, J., Stier, E., 2015. Applications of in vitro-in vivo correlations in generic drug development: case studies. AAPS. J 17, 1035-1039. Kesisoglou, F., Xia, B., Agrawal, N.G., 2015. Comparison of deconvolution-based and absorption modeling IVIVC for extended release formulations of a BCS III drug development candidate. AAPS. J 17, 1492-1500. Lamba, M., Wang, R., Fletcher, T., Alvey, C., Kushner, J., Stock, T.C., 2016. Extended-release once-daily formulation of tofacitinib: evaluation of pharmacokinetics compared with immediaterelease tofacitinib and impact of food. J. Clin. Pharmacol 56, 1362-1371. Malaterre, V., Ogorka, J., Loggia, N., Gurny, R., 2009. Oral osmotically driven systems: 30 years of development and clinical use. Eur. J. Pharm. Biopharm 73, 311-323. Pandey, P., Bindra, D.S., 2014. A commentary on scale-up of pan coating process using microenvironmental control. J Pharm Sci 103, 3412-3415. Pandey, P., Turton, R., Joshi, N., Hammerman, E., Ergun, J., 2006. Scale-up of a pan-coating process. AAPS PharmSciTech 7, 102.

33

Roudier, B., Davit, B.M., Beyssac, E., Cardot, J.M., 2014. In vitro- in vivo correlation's dissolution limits setting. Pharm. Res 31, 2529-2538. Schiller, C., Fröhlich, C.P., Giessmann, T., Siegmund, W., Mönnikes, H., Hosten, N., Weitschies, W., 2005. Intestinal fluid volumes and transit of dosage forms as assessed by magnetic resonance imaging. Aliment. Pharmacol. Ther 22, 971-979. Shamblin, S., 2010. Controlled release using bilayer osmotic tablet technology: reducing theory to practice, in: Wen, H., Park, K. (Eds.), Oral Controlled Release Formulation Design and Drug Delivery: Theory to Practice. John Wiley & Sons. Verma, R.K., Krishna, D.M., Garg, S., 2002. Formulation aspects in the development of osmotically controlled oral drug delivery systems. J. Control Release 79, 7-27. Waterman, K.C., MacDonald, B.C., Roy, M.C., 2009. Extrudable core system: development of a single-layer osmotic controlled-release tablet. J. Control Release 134, 201-206.

34

Tables Table 1 Composition and manufacturing scale for tofacitinib MR tablets. Description

11 mg medium-

11 mg fast-

11 mg slow-

release tablet

release tablet

release tablet

22 mg tablet

Tablet core Manufacturing-scale

Commercial-scale

Commercial-scale

Commercial-scale

Lab-scale

(300 kg)

(300 kg)

(300 kg)

(7 kg)

Tofacitinib citrate (mg)

17.771

17.771

17.771

35.541

Sorbitol (mg)

152.229

152.229

152.229

132.459

Hydroxyethyl cellulose (mg)

16.000

16.000

16.000

16.000

Copovidone (mg)

12.000

12.000

12.000

12.000

-

-

-

2.000

2.000

2.000

2.000

2.000

Colloidal silicon dioxide (mg) Magnesium stearate (mg)

Functional film coat Manufacturing-scale

Commercial-scale

Lab-scale

Lab-scale

Lab-scale

Cellulose acetate (mg)

9.840

6.240

9.120

7.200

Hydroxypropyl cellulose (mg)

6.560

4.160

6.080

4.800

Acetone (mg)

(423.120)a

(268.322)a

(392.160)a

(309.600)a

Methanol (mg)

(107.147)a

(67.947)a

(99.307)a

(78.400)a

Non-functional film coat Manufacturing-scale

Commercial-scale

Lab-scale

Lab-scale

Lab-scale

8.000

7.785

8.000

7.844

Purified water (mg)

(58.667)a

(57.089)a

(58.667)a

(57.523)a

Total (mg/tablet)

224.400

218.185

223.200

219.844

Opadry pink (mg)

a

Volatile component: removed during processing.

MR, modified-release.

35

Table 2 Summary of Weibull equation parameters for tofacitinib MR dissolution profiles Dosage Form

F

tlag (hr)a

MDT (hr)a

ba

int

R2

MR 11 fast

1

0.75 (4.2)

1.93 (2.2)

1.13 (2.7)

0

99.95%

MR 11 medium

1

0.89 (4.0)

2.23 (2.0)

1.17 (2.7)

0

99.94%

MR 11 slow

1

1.04 (4.5)

2.70 (2.2)

1.25 (2.9)

0

99.95%

MR 22

1

1.24 (3.5)

2.58 (2.7)

0.93 (3.4)

0

99.88%

a

Data reported as: parameter value (CV%).

b, slope factor; CV, coefficient of variation; F, amount released at time infinity; int, y-intercept; MDT, mean dissolution time; MR, modified-release; R2, coefficient of determination; t lag, lag time to onset of release.

36

Table 3 Descriptive summary of plasma tofacitinib in vivo PK parameter values

Parameter (unit) N

Parameter summary statisticsa by tofacitinib treatment MR 11 mg MR 11 mg MR 11 mg IR 5 mg MR 22 mg medium-release slow-release fast-release 36 36 36 36 36

AUCinf (ng•hr/ml)

300.7 (27)

294.9 (25)

310.0 (27)

152.9 (24)

599.0 (25)

Cmax (ng/ml)

41.40 (29)

37.23 (26)

44.83 (26)

48.14 (23)

76.47 (23)

Tmax (hr) t½ (hr) a

4.00 (2.00, 4.00) 4.00 (3.00, 6.00) 3.00 (2.00, 6.00) 0.50 (0.50, 2.00) 4.00 (2.00, 6.07) 5.535  2.740

5.869  2.618

5.838  2.914

3.322  0.466

6.437  2.798

Geometric mean (geometric %CV) for all except: median (range) for T max; arithmetic mean 

SD for t½. AUCinf, area under the curve extrapolated to time infinity; C max, maximum concentration; CV, coefficient of variation; IR, immediate-release; MR, modified-release; PK, pharmacokinetics; Tmax, time to reach maximum concentration; t 1/2, drug half-life.

37

Table 4 Statistical summary of treatment comparisons for plasma tofacitinib in vivo PK parameters Adjusted geometric means

Ratio (Test/Reference) of adjusted geometric Test Reference meansa MR 11 mg slow-release (Test) vs MR 11 mg medium-release (Reference) AUCinf (ng•hr/ml) 294.9 300.7 98.07 Parameter (unit)

Cmax (ng/ml)

37.23

94.44, 101.85

89.94

84.31, 95.94

MR 11 mg fast-release (Test) vs MR 11 mg medium-release (Reference) AUCinf (ng•hr/ml) 310.0 300.7 103.11

99.29, 107.08

Cmax (ng/ml)

108.30

101.52, 115.52

MR 11 mg fast-release (Test) vs MR 11 mg slow-release (Reference) AUCinf (ng•hr/ml) 310.0 294.9

105.14

101.24, 109.19

Cmax (ng/ml)

120.41

112.88, 128.45

MR 22 mg (Test) vs MR 11 mg medium-release (Reference) AUCinf, dn (ng•hr/ml)b 27.2 27.3

99.63

94.23, 105.34

Cmax,dn (ng/ml)b

92.36

86.21, 98.95

44.83

44.83

3.5

41.40

90% CI for ratio

41.40

37.23

3.8

MR 11 mg slow-release (Test) vs MR 22 mg (Reference) AUCinf, dn (ng•hr/ml)b

26.8

27.2

98.41

95.18, 101.76

Cmax,dn (ng/ml)b

3.4

3.5

97.40

91.81, 103.33

a

The ratios (and 90% CIs) are expressed as percentages.

b

Dose-normalized values.

AUCinf, area under the curve extrapolated to time infinity; Cmax, maximum concentration; CI, confidence interval; MR, modified-release.

38

Table 5 IVIVC piecewise (a) and simple linear (b) model correlation parameters a) Piecewise linear model MR 11 mg fast-, medium-, and MR 11 mg fast- and slow-release slow-release as internal validation as internal validation Model parameter AbsScale

b) Simple linear model MR 11 mg fast-, medium-, and MR 11 mg fast- and slow-release slow-release as internal validation as internal validation

Value (CV%)

95% CI

Value (CV%)

95% CI

Value (CV%)

95% CI

Value (CV%)

95% CI

0.954 (0.08)

0.952, 0.956

0.934 (0.12)

0.932, 0.936

0.906 (0.16)

0.903, 0.908

0.886 (0.15)

0.884, 0.889

t1 (hr)

5.20 (0.67)

5.14, 5.27

5.63 (0.68)

5.55, 5.70

-

-

-

-

timescale2

0.121 (2.14)

0.116, 0.127

0.127 (2.28)

0.122, 0.133

-

-

-

-

timescale1

0.80 (0.35)

0.799, 0.810

0.801 (0.34)

0.796, 0.807

0.790 (1.01)

0.775, 0.806

0.800 (0.96)

0.785, 0.815

tcut-off (hr)

24.5 (1.35)

23.8, 25.1

26.0 (1.35)

25.3, 26.7

-

-

-

-

CI, confidence interval; CV, coefficient of variation; IVIVC, in-vitro in-vivo correlation; MR, modified-release; t, time.

39

Table 6 IVIVC piecewise (a) and simple linear (b) model validation results with MR 11 mg fast-, medium-, and slow-release as internal validation, and MR 22 mg as external validation a) Piecewise linear model AUCinf (ng*hr/ml) Cmax (ng/ml) Formulation Geo.Mean Geo.Mean %PE Geo.Mean Geo.Mean observed predicted observed predicted MR 11 mg fast-, medium-, and slow-release as internal validation MR 11 mg fast- release 310.2 316.4 2.0 44.83 43.76 tablet MR 11 mg medium301.0 295.3 -1.9 41.40 41.13 release tablet MR 11 mg slow- release 294.9 300.4 1.9 37.23 36.74 tablet Average absolute 1.9 internal %PE MR 22 mg as external validation MR 22 mg 599.3 571.7 -4.6 76.47 71.70 tablet

%PE

b) Simple Linear Model AUCinf (ng*hr/ml) Cmax (ng/ml) Geo.Mean Geo.Mean %PE Geo.Mean Geo.Mean observed predicted observed predicted

-2.4

310.2

307.6

-0.9

44.83

41.19

-8.1

-0.7

301.0

287.3

-4.5

41.40

38.69

-6.6

-1.3

294.9

307.5

4.3

37.23

34.91

-6.2

1.5

-

-

3.2

-

-

7.0

-6.2

599.3

603.1

0.6

76.47

67.55

-11.7

%PE, prediction error; AUCinf, area under the curve extrapolated to time infinity; Cmax, maximum concentration; GeoMean, geometric mean; IVIVC, in-vitro in-vivo correlation; MR, modified-release.

40

%PE

Table 7 IVIVC piecewise (a) and simple linear (b) model validation results with MR 11 mg fast- and slow-release as internal validation, and MR 11 mg medium-release and MR 22 mg as external validation a) Piecewise model AUCinf (ng*hr/ml) Formulation Geo.Mean Geo.Mean %PE observed predicted MR 11 mg fast- and slow-release as internal validation

Cmax (ng/ml) Geo.Mean Geo.Mean observed predicted

MR 11 mg fast301.2 313.3 1.0 release tablet MR 11 mg slow294.9 302.2 2.5 release tablet Average absolute 1.7 internal %PE MR 11 mg medium-release and MR 22 mg as external validation MR 11 mg medium301.0 291.0 -3.3 release tablet MR 22 mg tablet 599.3 575.9 -3.9

%PE

b) Simple linear model AUCinf (ng*hr/ml) Cmax (ng/ml) Geo.Mean Geo.Mean %PE Geo.Mean Geo.Mean observed predicted observed predicted

%PE

44.83

42.78

-4.6

310.2

301.2

-2.9

44.83

40.55

-9.5

37.23

36.00

-3.3

294.9

301.1

2.1

37.23

34.44

-7.5

-

-

3.9

-

-

2.5

-

-

8.5

41.40

40.19

-2.9

301.0

280.8

-6.7

41.40

38.11

-7.9

76.47

70.07

-8.4

599.3

591.1

-1.4

76.47

66.60

-12.9

%PE, prediction error; AUCinf, area under the curve extrapolated to time infinity; C max, maximum concentration; GeoMean, geometric mean; IVIVC, in-vitro in-vivo correlation; MR, modified-release.

41

Figure legends

Fig. 1. Manufacturing process diagram. MR, modified-release

Fig. 2. Mean (SD) in vitro dissolution profiles for tofacitinib MR tablets. MR, modified-release; SD, standard deviation 42

Fig. 3. Observed and model-fitted in vitro dissolution data using the Weibull equation. MR, modified-release.

43

Fig. 4. Median (SD) plasma tofacitinib in vivo concentration-time profiles following single oral doses. The lower level of quantification was 0.100 ng/ml. Summary statistics were calculated by setting concentration values below the lower limit to 0. IR, immediate-release; MR, modified-release; SD, standard deviation

44

Fig. 5. Mean in vivo absorption profiles for tofacitinib MR 11 mg and MR 22 mg tablets. MR, modified-release.

45

Fig. 6. In vivo absorption versus in vitro dissolution relationship for tofacitinib MR tablets. MR, modified-release.

46

Fig. 7. Comparison of observed and user-built piecewise IVIVC predicted mean PK profiles for tofacitinib MR tablets. A) MR 11 mg fast-release, B) MR 11 mg slow-release, C) MR 11 mg medium-release, and D) MR 22 mg. IVIVC, in-vitro in-vivo correlation; MR, modified-release; PK, pharmacokinetic.

47

GRAPHICAL ABSTRACT

48