Bioequivalence and food effect of heat-stressed and non⿿heat-stressed dapagliflozin 2.5- and 10-mg tablets

Bioequivalence and food effect of heat-stressed and non⿿heat-stressed dapagliflozin 2.5- and 10-mg tablets

Accepted Manuscript Title: Bioequivalence and food effect of heat-stressed and non–heat-stressed dapagliflozin 2.5- and 10-mg tablets Author: Frank La...

1MB Sizes 0 Downloads 115 Views

Accepted Manuscript Title: Bioequivalence and food effect of heat-stressed and non–heat-stressed dapagliflozin 2.5- and 10-mg tablets Author: Frank LaCreta Steven C. Griffen Xiaoni Liu Charles Smith Carey Hines Kevin Volk Ravindra Tejwani David W. Boulton PII: DOI: Reference:

S0378-5173(16)30647-0 http://dx.doi.org/doi:10.1016/j.ijpharm.2016.07.017 IJP 15914

To appear in:

International Journal of Pharmaceutics

Received date: Revised date: Accepted date:

7-1-2016 5-7-2016 9-7-2016

Please cite this article as: LaCreta, Frank, Griffen, Steven C., Liu, Xiaoni, Smith, Charles, Hines, Carey, Volk, Kevin, Tejwani, Ravindra, Boulton, David W., Bioequivalence and food effect of heat-stressed and non–heat-stressed dapagliflozin 2.5- and 10-mg tablets.International Journal of Pharmaceutics http://dx.doi.org/10.1016/j.ijpharm.2016.07.017 This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. 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.

Bioequivalence and food effect of heat-stressed and non–heat-stressed dapagliflozin 2.5- and 10-mg tablets Frank LaCretaa, Steven C. Griffena,1, Xiaoni Liub, Charles Smithc, Carey Hinesd, Kevin Volke, Ravindra Tejwanie,2, David W. Boultonf,* a

Bristol-Myers Squibb Company, Route 206 and Province Line Road, Princeton, NJ 08543,

USA; [email protected] b

Bristol-Myers Squibb Company, Route 206 and Province Line Road, Princeton, NJ 08543,

USA; [email protected] c

Pharmaceutical Product Development, 5516 Falmouth Street, Suite 301 ABC, Richmond, VA

23230, USA; [email protected] d

Pharmaceutical Product Development, 5516 Falmouth Street, Suite 301 ABC, Richmond, VA

23230, USA; [email protected] e

Bristol-Myers Squibb Company, 1 Squibb Drive, New Brunswick, NJ 08901, USA;

[email protected] f

AstraZeneca, One MedImmune Way, Gaithersburg, MD 20878, USA;

[email protected]

* Corresponding author at One MedImmune Way, Gaithersburg, MD 20878, USA

Tel: +1-301-398-5564. E-mail address: [email protected] (D. Boulton). 1

Current affiliation: JDRF, 26 Broadway, 14th Floor, New York, NY 10004, USA;

[email protected] 1

2

Current affiliation: ACADIA Pharmaceuticals, 3611 Valley Centre Drive, San Diego, CA 92130-

3331, USA; [email protected]

2

Graphical abstract

3

ABSTRACT Physical storage of formulations may result in physical composition changes that affect pharmacokinetics. Dapagliflozin, an oral sodium-glucose cotransporter 2 inhibitor used for type 2 diabetes mellitus, stored under prolonged exposure to heat converts crystalline dapagliflozin to an amorphous form. Bioequivalence of the amorphous to crystalline form and food effects of each form in the 2.5-mg formulation are unknown. Two open-label, crossover, single-dose studies in healthy participants assessed pharmacokinetics for heat-stressed (HS) and non–heatstressed (NH) dapagliflozin 10-mg (study 1, N=29, fasted + HS food effect) and 2.5-mg (study 2, N=28, fasted + HS and NH food effect) tablets. The 90% confidence intervals for geometric mean ratios of area under the concentration–time curve (AUC) and peak concentration (Cmax) for HS 2.5- and 10-mg tablets were within 80–125%, indicating bioequivalence. In the fed vs. fasted state for 2.5-mg and 10-mg HS tablets, AUCs were similar, time to Cmax was prolonged by 1.25 hours, and Cmax decreased by approximately 50%. No serious adverse events were reported. Given that dapagliflozin’s efficacy is dependent upon AUC, it was concluded that HS and NH dapagliflozin tablets are bioequivalent in 2.5- and 10-mg doses with no clinically meaningful food effect for either form.

Keywords: Bioequivalence; Dapagliflozin; Pharmacokinetics; Sodium-glucose cotransporter 2 inhibitor; Drug stability; Heat-stressed; Amorphous form

4

Abbreviations AUC0–t, area under the plasma concentration–time curve from time 0 to the last quantifiable concentration AUC0–inf, area under the plasma concentration–time curve from time 0 to infinity Cp, plasma concentration Cmax, maximum Cp Dapagliflozin, [2S,3R,4R,5S,6R]-2-[4-chloro-3-(4-ethoxybenzyl)phenyl]-6(hydroxymethyl)tetrahydro-2H-pyran-3,4,5-triol, compounded with (2S)-1,2-propanediol, hydrate (1:1:1) HS, heat-stressed Kel, terminal rate constant LS, least squares NH, non–heat-stressed tmax, time to Cmax t1/2, terminal half-life

5

1. Introduction Stability testing of pharmaceutical products is critical to the drug development process as it helps ensure the identity, potency, and purity of the drug components, and those of the formulated product as a whole. It also supports patient safety, as medication instability may have negative effects on bioavailability, safety, and therapeutic efficacy (Bajaj et al., 2012; US Food and Drug Administration et al., 2003). Stability testing includes storage at elevated temperatures and humidity levels for short periods, which simulates the effects of storage under normal conditions for a long duration of time (Bajaj et al., 2012; European Medicines Agency, 2000; US Food and Drug Administration et al., 2003). Dapagliflozin, a highly selective, orally active, reversible inhibitor of human sodium-glucose cotransporter 2 (AstraZeneca, 2015; Meng et al., 2008), is indicated as an adjunct to diet and exercise for the treatment of adults with type 2 diabetes mellitus (AstraZeneca, 2015). Dapagliflozin ([2S,3R,4R,5S,6R]-2-[4-chloro-3-(4-ethoxybenzyl)phenyl]-6(hydroxymethyl)tetrahydro-2H-pyran-3,4,5-triol), compounded with (2S)-1,2-propanediol, hydrate (1:1:1), is known to be converted by heat stress from the crystalline to the amorphous form (European Medicines Agency, 2012). Studies on characterization of dapagliflozin solvate crystals have indicated that complete conversion to the amorphous form occurs almost instantly at 50°C and practically irreversibly, given the loss of water and propanediol from the dapagliflozin solvate crystal that occurs during the process (data on file). While the conversion of dapagliflozin to the amorphous form may occur during storage, it is not known whether this change affects the pharmacokinetics (PK) of dapagliflozin, as may be expected with some polymorphic changes (US Food and Drug Administration, Center for Drug Evaluation and Research, 2007). This is because a drug that exists in various polymorphic forms can have different aqueous solubility and dissolution rates for each form (US Food and

6

Drug Administration, Center for Drug Evaluation and Research, 2007). Even though dapagliflozin tablets are manufactured using the crystalline drug form, it was considered possible that product aging could potentially lead to amorphous conversion under the recommended storage conditions, and that a mixture of crystalline and amorphous drug forms may be present in the drug product toward the end of its shelf life. A risk assessment (based on the International Conference on Harmonisation Q 6 A) (European Medicines Agency, 2000) was therefore conducted, and investigational studies (both in vitro and in vivo) were initiated to evaluate the significance of this form change. Despite conversion to the amorphous form, no chemical degradation of dapagliflozin, nor any changes in dapagliflozin assay, were observed after the tablets were heat stressed (European Medicines Agency, 2012). Aqueous in vitro dissolution tests (Supplementary Table) indicated that there was no difference in the rate and extent of release and dissolution of dapagliflozin from non–heat-stressed (NH) dapagliflozin 2.5- and 10-mg tablets (crystalline dapagliflozin) and heat-stressed (HS) dapagliflozin 2.5- and 10-mg tablets (amorphous dapagliflozin) (European Medicines Agency, 2012). However, it was not known what other effects heat stress may have on formulation performance. In addition to stability testing, it is also important to determine any potential impact that food may have on the PK of new medicines, as this will influence clinical use with respect to timing of drug administration relative to meals. The effect of food on the PK of medicines is variable, not entirely predictable, and often dependent upon the physiochemical and gastrointestinal permeability characteristics of the drug (US Food and Drug Administration, Center for Drug Evaluation and Research, 2002). Therefore, it may be important to assess the impact of food on the rate and extent of drug absorption.

7

A previous investigation in healthy participants indicated that food does not affect the overall extent of absorption or the metabolism of orally administered, NH 10-mg dapagliflozin tablets (Kasichayanula et al., 2011). Participants who received dapagliflozin following a high-fat meal had comparable systemic exposure to participants treated in the fasted state, as assessed by area under the plasma concentration–time curve (AUC). This is in agreement with the high aqueous solubility of dapagliflozin (>1 mg/mL at physiological pH) and high degree of intestinal permeability (Kasichayanula et al., 2011). These participants did have a lower mean maximum plasma concentration (Cmax) for dapagliflozin and an increased time to Cmax (tmax), consistent with a delay in the rate of gastric emptying following ingestion of a meal. However, an earlier analysis demonstrated that similar variations in Cmax and Tmax following peri-prandial administration of dapagliflozin (2.5–10 mg/day) do not affect the cumulative amount of glucose excreted (Komoroski et al., 2009). Furthermore, in phase 3 trials, patients with type 2 diabetes mellitus showed significant improvements in glycemic control after 24 weeks of receiving dapagliflozin irrespective of meal times (Bailey et al., 2010; Ferrannini et al., 2010). Considering these findings together, it has been concluded that the effects of food on the rate of absorption of dapagliflozin are not clinically meaningful and that the overall level of dapagliflozin-induced glucose excretion is dependent upon dapagliflozin AUC (Kasichayanula et al., 2011). Even so, at present, the effects of food on HS dapagliflozin and NH 2.5-mg tablets are unknown. Thus, it was considered important to assess the potential effects of the dapagliflozin form change. This would help to validate a proposed product shelf life of at least 3 years, by allowing for a near-complete change from crystalline to amorphous dapagliflozin. This study in healthy participants was undertaken to evaluate the fasted-state bioequivalence of NH and HS dapagliflozin tablets at 2.5- and 10-mg doses and to evaluate the effect of a high-fat, highcalorie meal on the PK of HS dapagliflozin tablets at 2.5- and 10-mg doses.

2. Methods 8

2.1. Study design HS dapagliflozin tablets were exposed to 50°C, ambient humidity for 6 weeks. Owing to differences in the formulation of the dapagliflozin 2.5- and 10-mg tablets, two studies were conducted. Study 1 examined the fasted-state bioequivalence of NH and HS dapagliflozin 10-mg tablets and the effect of a high-fat, high-calorie meal on the PK of HS dapagliflozin 10-mg tablets. Study 2 examined fasted-state bioequivalence of NH and HS dapagliflozin 2.5-mg tablets and evaluated the effects of food on PK for both NH and HS 2.5-mg tablets. Study 1 did not evaluate food effects for NH dapagliflozin 10-mg tablets as this has already been characterized (Kasichayanula et al., 2011). Healthy participants aged 18–55 years with a body mass index of 18–32 kg/m2 were eligible for inclusion. Participants were excluded if they had any significant acute or chronic medical illness or evidence of organ dysfunction, or any clinically significant abnormalities in their medical history, physical examination, and/or clinical laboratory determinations. All participants underwent eligibility screening within 21 days before study drug administration on day 1 of period 1. Study 1 was an open-label, randomized, three-period, three-treatment, crossover, single-dose study in fasted and fed healthy participants (Fig. 1A), which took place between 20 April and 26 May 2010. Participants were randomly assigned to one of six treatment sequences (ABC, BCA, CAB, CBA, ACB, or BAC), where A = NH dapagliflozin 10-mg tablet administered under fasted conditions, B = HS dapagliflozin 10-mg tablet administered under fasted conditions, and C = HS dapagliflozin 10-mg tablet administered after a high-fat meal. Study 2 was an open-label, randomized, four-period, four-treatment, crossover, single-dose study in fasted and fed healthy participants (Fig. 1B), which took place between 10 July and 13 August 2010. Participants were randomly assigned to one of four treatment sequences (DGEF, EDFG, FEGD, GFDE), where D = NH dapagliflozin 2.5-mg tablet administered under fasted 9

conditions, E = HS dapagliflozin 2.5-mg tablet administered under fasted conditions, F = NH dapagliflozin 2.5-mg tablet administered after a high-fat meal, and G = HS dapagliflozin 2.5-mg tablet administered after a high-fat meal. For both studies, participants received each treatment once (one treatment per period). Participants receiving treatments A, B, D, and E had nothing to eat or drink except water from 10 hours before until 4 hours after treatment administration; for 1 hour before and after treatment, water was only allowed with dosing. The standard high-fat, high-calorie breakfast (937 total calories: 13% protein, 54% fat, and 33% carbohydrates) given to participants receiving treatments C, F, and G, was consumed 30 minutes before dosing. The meal consisted of two eggs fried in 2 teaspoons of butter, three strips of bacon, two slices of white toast with 1 teaspoon of butter, 1 tablespoon of jam, 4 ounces of hash brown potatoes cooked with 2 teaspoons of butter, and 8 fluid ounces of whole milk. After each treatment, participants had a minimum 4-day washout prior to the next treatment. Serial blood samples for PK parameter evaluation of dapagliflozin in plasma were collected before dosing and over 48 hours (study 1) or 24 hours (study 2) after dosing in each period. Both studies were performed in accordance with the Declaration of Helsinki and consistent with the International Conference on Harmonisation’s Good Clinical Practice guidelines and applicable regulatory requirements. All participants gave informed written consent prior to study participation. 2.2. Primary objectives The primary objectives of these two studies were to assess the fasted-state bioequivalence of a HS dapagliflozin 10-mg tablet vs. a NH 10-mg tablet (study 1) and to assess the fasted-state bioequivalence of a HS dapagliflozin 2.5-mg tablet vs. a NH 2.5-mg tablet (study 2). 2.3. Secondary objectives

10

The secondary objectives of these two studies were to characterize the effects of a high-fat, high-calorie meal on the PK of a HS dapagliflozin 10-mg tablet compared with fasted conditions (study 1) and to characterize the effects of a high-fat, high-calorie meal on the PK of a HS dapagliflozin 2.5-mg tablet and a NH 2.5-mg tablet compared with fasted conditions (study 2). In addition, the safety and tolerability of dapagliflozin following oral administration were investigated. 2.4. Pharmacokinetic evaluation In order to determine the plasma concentrations (Cp) of dapagliflozin, blood samples were collected before dosing and at 0.25, 0.5, 0.75, 1, 1.5, 2, 4, 8, 12, 16, and 24 hours after dosing with a 2.5- or 10-mg dapagliflozin tablet; in addition, blood samples were collected at 36 and 48 hours only after dosing with a 10-mg dapagliflozin tablet. A validated high-performance liquid chromatography tandem mass spectrometry method was used to determine Cp. Dapagliflozin PK parameters were derived from Cp vs. time data using noncompartmental methods and actual blood sampling times. Evaluated parameters included AUC from time 0 to the last quantifiable concentration (AUC0–t), AUC from time 0 to infinity (AUC0–inf), Cmax, tmax, and terminal half-life (t1/2). Both AUC0–inf and t1/2 were calculated using the terminal rate constant (Kel). The bioequivalence of NH and HS dapagliflozin tablets at 2.5- and 10-mg doses was assessed by comparing geometric least squares (LS) mean ratios for the fasted-state HS treatment (B, study 1; E, study 2) vs. the fasted-state NH treatment (A, study 1; D, study 2). In study 1, the effect of food on PK for HS dapagliflozin 10-mg tablets was determined by comparing the fed-state HS treatment (C) with the fasted-state HS treatment (B). In study 2, the effect of food on the PK of dapagliflozin 2.5-mg tablets was assessed by comparing the fedstate NH treatment (F) with the fasted-state NH treatment (D) and comparing the fed-state HS treatment (G) with the fasted-state HS treatment (E). 11

2.5. Safety assessments Safety assessments included adverse events (AEs), clinical laboratory results, vital sign measurements, 12-lead electrocardiogram results, and physical examination findings. 2.6. Statistical methods All analyses were performed using SAS software (SAS Institute, Cary, NC, USA), version 8.2 or higher. Descriptive statistics for continuous measures included the number of participants, mean, standard deviation, coefficient of variation, median, and range. The geometric LS means for Cmax, AUC0–t, and AUC0–inf were used to assess bioequivalence and any potential food effects. Bioequivalence and absence of a food effect were concluded if the 90% confidence intervals (CIs) for the geometric LS mean ratios for the relevant treatment arm comparisons were within 80–125% for both Cmax and AUC0–inf. Assuming no difference in bioavailability between HS and NH dapagliflozin tablets, it was determined that data from 24 subjects were needed to provide 96% and 99% power to conclude bioequivalence with respect to Cmax and AUC0–inf, respectively. If there was a 5% difference in bioavailability, 24 subjects would be needed to provide 88% (Cmax) and 99% (AUC0–inf) power to conclude bioequivalence between HS and NH tablets. It was also calculated that data from 24 participants were needed to provide at least 90% confidence that the estimates of the fed-tofasted ratios of geometric LS means were within 10% of the true value for dapagliflozin Cmax and within 5% for dapagliflozin AUC0–inf. Moreover, data from 24 participants would have provided at least 99% power to conclude the absence of a food effect on AUC0–inf of dapagliflozin. Point estimates and 90% CIs were constructed from fitting the general linear mixed models for log-transformed data, with treatment and period as fixed effects and measurements within participants as repeated measurements. No adjustment was made for multiplicity.

12

3. Results 3.1. Participant demographics A total of 29 participants were enrolled in study 1 and 28 (96.6%) completed the study; one participant discontinued for nonmedical reasons. In study 2, a total of 28 participants were enrolled and 25 participants (89.3%) completed the study; one participant discontinued due to pregnancy and two participants discontinued due to nonmedical reasons. No dropout participants were replaced in either study. Demographic and baseline characteristics were similar between the studies (Table 1). 3.2. Pharmacokinetic comparison of HS vs. NH dapagliflozin tablets in the fasted and fed states Plasma PK parameters for dapagliflozin 10- and 2.5-mg tablets are summarized in Tables 2 and 3, respectively; statistical comparisons are summarized in Tables 4 and 5. Heat stress had no effect on total exposure in study 1. For participants receiving dapagliflozin HS and NH 10-mg tablets, the geometric LS mean systemic exposure (AUC0–inf and AUC0–t) and Cmax were highly comparable for both forms under fasting conditions (Table 2). Median tmax (0.75 and 1.00 hours, respectively) and mean t1/2 (13.1 and 11.6 hours, respectively) were also similar between these treatment groups. Furthermore, the 90% CIs of the geometric LS mean ratios for Cmax, AUC0–inf, and AUC0–t were completely contained within the 80–125% limits for concluding the bioequivalence of HS and NH dapagliflozin 10-mg tablets when administered under fasting conditions (Table 4). Food also had no effect on total exposure in study 1, as measured by AUC0–inf or t1/2 (Table 2). The 90% CIs for the ratios of AUC0–t and AUC0–inf were completely contained within the 80– 125% criterion limits when HS dapagliflozin 10-mg tablets were administered under fed vs. fasting conditions, indicating no food effect on total systemic exposure of dapagliflozin (Table 4).

13

However, there was a 1.25-hour delay in the tmax for HS dapagliflozin 10-mg tablets administered under fed vs. fasting conditions (Fig. 2A). This resulted in a lower Cmax (~ 50% decrease) with 90% CIs for the comparison ratio of geometric LS means that were completely outside the 80–125% criterion limit (Table 4). Similarly to study 1, heat stress had no effect on total exposure in study 2. Geometric mean exposures (AUC0–inf or AUC0–t) and geometric mean peak exposure (Cmax) were nearly identical for the HS and NH dapagliflozin 2.5-mg tablets administered under fasting conditions (Table 3 and Fig. 2B). Statistical comparison of geometric LS means for both sets of parameters between these treatment groups produced 90% CIs that were completely contained within the bioequivalence criterion interval of 80–125% (Table 5). As in study 1, food had no effect on total exposure in study 2. Participants who received NH or HS dapagliflozin 2.5-mg tablets under fed conditions produced highly comparable, bioequivalent geometric mean AUC0–inf and AUC0–t to participants receiving the same formulations under fasting conditions (Tables 3 and 5, and Fig. 2B). However, geometric mean Cmax was markedly reduced (by ~50%) under fed conditions while median tmax was delayed to 2.00 hours compared with 0.75 hours after dosing under fasting conditions. When comparing these participants (receiving dapagliflozin 2.5 mg under fed vs. fasting conditions), the 90% CIs of the ratio of geometric LS means were completely outside the 80–125% criterion limits (Table 5). Mean terminal elimination phase, as assessed by t1/2, was close to 10 hours for all treatments in study 2 (Table 3). 3.3. Safety evaluation In general, NH and HS 2.5- and 10-mg dapagliflozin tablets were safe and well tolerated in the fed and fasted states; there were no deaths, serious AEs, or AEs that led to study drug discontinuation in either study. A summary of AEs is available in Table 6. 14

4. Discussion Following the discovery that prolonged heat stress (50°C for ~1 month) changes dapagliflozin from a crystalline to an amorphous form, two studies investigated the potential impact of this change on the bioequivalence of NH and HS dapagliflozin 2.5- and 10-mg tablets. The findings of these studies indicate no differences in bioavailability between the amorphous and crystalline forms of dapagliflozin and no impact on in vivo product performance. Additionally, these studies evaluated the effect of food on NH and HS dapagliflozin 2.5-mg tablets and on HS dapagliflozin 10-mg tablets, and found no clinically meaningful food effects on the PK profiles of the NH and HS dapagliflozin tablets because the overall extent of exposure (as assessed by AUC) was unaffected by food. These conclusions are also relevant for similarly formulated doses of dapagliflozin between 2.5 and 10 mg that have not been studied, such as the marketed 5-mg dapagliflozin tablet (AstraZeneca, 2015). Sensitivity of drug activity to changing physical conditions is often affected by the presence of polymorphic conformers of the same drug (Savjani et al., 2012). These different forms may vary in physiochemical properties such as solubility and melting point. If the most thermodynamically stable form of a drug in a given environment confers lower solubility, this will likely reduce the rate of drug dissolution and its bioavailability in the body (Savjani et al., 2012). This is because after ingestion, an oral drug must dissolve into solution at the site of absorption before in can enter the systemic circulation for distribution to its site of action. Therefore, a thorough understanding of the physiochemical properties for all forms of a given drug is essential for considering the impact of physical conditions on the clinical safety and efficacy of the drug (European Medicines Agency, 2000). In these two studies, analysis of the food effect on HS dapagliflozin 2.5- and 10-mg tablets showed that there was no impact of a high-fat meal on the total systemic exposure of

15

dapagliflozin, although there was a ~50% decrease in Cmax and a prolonged tmax. Similar findings were observed for the NH dapagliflozin 2.5-mg tablets and have been previously reported for NH dapagliflozin 10-mg tablets (Kasichayanula et al., 2011). While no formal statistical testing comparing NH and HS dapagliflozin in the fed state was conducted, the magnitude of the food effect appeared to be similar in both forms. Although peak exposures were reduced and tmax prolonged for NH and HS dapagliflozin 2.5- and 10-mg tablets in the fed state vs. fasted state, these food effects are not expected to influence clinical efficacy and outcomes of dapagliflozin treatment. This is because dapagliflozin efficacy is dependent on total exposure (AUC0–inf) (Kasichayanula et al., 2011), which was not affected. Therefore, the Cmax reduction is not considered to be clinically meaningful. When administered as single oral doses of HS or NH tablets in both studies, dapagliflozin was well tolerated under fasted or fed conditions. In addition, there were no deaths, serious AEs, or AEs that led to study drug discontinuation. Thus, the dapagliflozin form change had no apparent effect on the drug’s safety profile, albeit from a single-dose experience. Since drug safety, performance, and efficacy are not affected after conversion from the crystalline to amorphous form, an acceptance criterion for polymorph content in the formulated drug was not set. This is an important outcome for establishing product shelf life because the stable performance of dapagliflozin, regardless of drug form, will allow for drug storage under less restrictive conditions while ensuring product quality. An important limitation of the methodology is that there was no direct analysis of the crystalline and amorphous state of the drug substance within the drug product. Because the drug product contains a very small amount of drug substance diluted with inactive ingredients, it is not possible to quantify crystalline or amorphous forms in the drug product with analytical methods such as X-ray diffraction and differential scanning calorimetry as can be applied to the drug

16

substance. However, as noted in the introduction, the drug substance converts from the crystalline form to the amorphous form almost instantly and irreversibly when exposed to 50°C temperature.

5. Key conclusions The data presented here show no statistically significant or clinically relevant difference in bioavailability between NH and HS dapagliflozin tablets when given under fasted conditions; in vivo product performance was not affected, irrespective of whether the administered tablets contained the amorphous or crystalline form of dapagliflozin. Furthermore, the total systemic exposure of dapagliflozin was not affected when the HS dapagliflozin 10-mg tablet was administered under fed conditions. Overall, dapagliflozin appears to be well tolerated as crystalline or amorphous forms under both fasted and fed conditions. Based on the findings from these two studies, no special storage conditions or specific changes to the prescribing information for administration of dapagliflozin are required. This should facilitate medication compliance by allowing administration under both fed and fasted conditions, and allow storage under less restrictive conditions. Conflict of interest Steven C. Griffen was an employee of Bristol-Myers Squibb when this research was performed, and is a current employee of JDRF; Charles Smith and Carey Hines are employees of PPD; Xiaoni Liu, Kevin Volk, and Frank LaCreta are employees/shareholders of Bristol-Myers Squibb; David W. Boulton is an employee/shareholder of AstraZeneca and a shareholder of BristolMyers Squibb; Ravindra Tejwani was an employee/shareholder of Bristol-Myers Squibb when

17

this research was performed, and is a current employee/shareholder of ACADIA Pharmaceuticals. Acknowledgements Editorial support was provided by Robert Axford-Gatley, MD, and Safeer Mughal, PhD, of PPSI (a PAREXEL company) and was funded by AstraZeneca. Role of the funding source These studies were funded by Bristol-Myers Squibb and AstraZeneca. The authors are employees or former employees of both companies and contributed to the study design, analysis and interpretation of data, writing of the report, and the decision to submit the article for publication. Contributor statements All authors contributed equally to the development this manuscript.

18

References

AstraZeneca 2015, Farziga (dapagliflozin) tablets [prescribing information], Wilmington, DE, USA. Available at: http://www.azpicentral.com/farxiga/pi_farxiga.pdf#page=1. Bailey, C.J., Gross, J.L., Pieters, A., Bastien, A., List, J.F., 2010. Effect of dapagliflozin in patients with type 2 diabetes who have inadequate glycaemic control with metformin: a randomised, double-blind, placebo-controlled trial. Lancet 375, 2223-2233. Bajaj, S., Singla, D., Sakhuja, N., 2012. Stability testing of pharmaceutical products. J. Appl. Pharm. Sci. 2, 129-138. European Medicines Agency. ICH Topic Q 6 A. Note for guidance specifications: test procedures and acceptance criteria for new drug substances and new drug products: chemical substances (CPMP/ICH/367/96). EMA Website. Available at: http://www.ema.europa.eu/docs/en_GB/document_library/Scientific_guideline/2009/09/WC5000 02823.pdf. Accessed on October 22, 2014. European Medicines Agency. Assessment report: Forxiga. EMA/689976/2012. EMA Website. Available at: http://www.ema.europa.eu/docs/en_GB/document_library/EPAR__Public_assessment_report/human/002322/WC500136024.pdf. Accessed on October 22, 2014. Ferrannini, E., Ramos, S.J., Salsali, A., Tang, W., List, J.F., 2010. Dapagliflozin monotherapy in type 2 diabetic patients with inadequate glycemic control by diet and exercise: a randomized, double-blind, placebo-controlled, phase 3 trial. Diabetes Care 33, 2217-2224. Kasichayanula, S., Liu, X., Zhang, W., Pfister, M., Reele, S.B., Aubry, A.F., LaCreta, F.P., Boulton, D.W., 2011. Effect of a high-fat meal on the pharmacokinetics of dapagliflozin, a selective SGLT2 inhibitor, in healthy subjects. Diabetes Obes. Metab. 13, 770-773. Komoroski, B., Vachharajani, N., Boulton, D., Kornhauser, D., Geraldes, M., Li, L., Pfister, M., 2009. Dapagliflozin, a novel SGLT2 inhibitor, induces dose-dependent glucosuria in healthy subjects. Clin. Pharmacol. Ther. 85, 520-526. Meng, W., Ellsworth, B.A., Nirschl, A.A., McCann, P.J., Patel, M., Girotra, R.N., Wu, G., Sher, P.M., Morrison, E.P., Biller, S.A., Zahler, R., Deshpande, P.P., Pullockaran, A., Hagan, D.L., Morgan, N., Taylor, J.R., Obermeier, M.T., Humphreys, W.G., Khanna, A., Discenza, L., Robertson, J.G., Wang, A., Han, S., Wetterau, J.R., Janovitz, E.B., Flint, O.P., Whaley, J.M., Washburn, W.N., 2008. Discovery of dapagliflozin: a potent, selective renal sodium-dependent glucose cotransporter 2 (SGLT2) inhibitor for the treatment of type 2 diabetes. J. Med. Chem. 51, 1145-1149. Savjani, K.T., Gajjar, A.K., Savjani, J.K., 2012. Drug solubility: importance and enhancement techniques. ISRN. Pharm. 2012, 195727. US Food and Drug Administration & Center for Drug Evaluation and Research 2002, Guidance for industry. Food-effect bioavailability and fed bioequivalence studies, US FDA, Rockville, MD.

19

US Food and Drug Administration & Center for Drug Evaluation and Research 2007, Guidance for industry. ANDAs: pharmaceutical solid polymorphism. Chemistry, manufacturing, and controls information, US FDA, Rockville, MD. US Food and Drug Administration, Center for Drug Evaluation and Research, & Center for Biologics Evaluation and Research 2003, Guidance for industry. Q1A(R2) stability testing of new drug substances and products, US FDA, Rockville, MD.

20

Table 1. Summary of participant demographics and baseline characteristics. Parameter

Study 1

Study 2

Dapagliflozin 10-mg tablets

Dapagliflozin 2.5-mg tablets

(N = 29)

(N = 28)

34.7 (10.5)

32.3 (9.2)

33.0

31.0

19, 54

20, 51

Males, n (%)

13 (44.8)

10 (35.7)

Females, n (%)

16 (55.2)

18 (64.3)

28 (96.6)

23 (82.1)

0

4 (14.3)

1 (3.4)

1 (3.6)

Hispanic or Latino

10 (34.5)

12 (42.9)

Not Hispanic or Latino

19 (65.5)

16 (57.1)

170.8 (10.9)

163.3 (9.9)

173.0

163.9

149.2, 186.8

145.4, 182.8

73.5 (12.5)

70.1 (11.6)

74.9

67.1

50.0, 95.6

53.1, 92.9

Age, years Mean (SD) Median Minimum, maximum

Race, n (%) Caucasian Black or African American Asian Ethnicity, n (%)

Height, cm Mean (SD) Median Minimum, maximum Weight, kg Mean (SD) Median Minimum, maximum

21

Body mass index, kg/m2 Mean (SD) Median Minimum, maximum

25.0 (2.5)

26.1 (2.5)

24.2

26.4

22.0, 31.2

21.5, 32.0

Percentages were based on the overall number of participants in the safety population. SD, standard deviation.

22

Table 2. Plasma pharmacokinetic parameters of dapagliflozin 10-mg tablets. Treatment Parameter

NH

HS

HS

dapagliflozin 10 mg,

dapagliflozin 10 mg,

dapagliflozin 10 mg,

fasting

fasting

fed

(N = 28)

(N = 29)

(N = 29)

AUC0–t, ng·h/mL

637 (25)

629 (25)

604 (25)

AUC0–inf, ng·h/mLa

663 (25)

650 (24)

635 (25)

Cmax, ng/mL

172 (32)

176 (30)

97 (28)

tmax, h

1.00 (0.75–2.00)

0.75 (0.50–1.50)

2.00 (0.75–4.00)

t1/2, ha

11.6 (3.5)

13.1 (4.2)

13.2 (3.8)

Note: Geometric mean (arithmetic mean CV%) is reported only for AUCs and Cmax. Median (range) is presented for tmax. Arithmetic mean (SD) is presented for t1/2. a

The sample sizes for these parameters were N = 28 for all treatments because two profiles did

not have sufficiently well-defined Kel to include in the analysis. AUC0–inf, area under the plasma concentration–time curve from time 0 to infinity; AUC0–t, area under the plasma concentration–time curve from time 0 to the last quantifiable concentration; Cmax, maximum plasma concentration; CV, coefficient of variation; HS, heat-stressed; Kel, terminal rate constant; NH, non–heat-stressed; SD, standard deviation; tmax, time to Cmax; t1/2, drug terminal half-life.

23

Table 3. Plasma pharmacokinetic parameters of dapagliflozin 2.5-mg tablets. Treatment Parameter

NH

HS

NH

HS

dapagliflozin 2.5 mg,

dapagliflozin 2.5 mg,

dapagliflozin 2.5 mg,

dapagliflozin 2.5 mg,

fasting

fasting

fed

fed

(N = 28)

(N = 26)

(N = 27)

(N = 28)

AUC0–t, ng·h/mL

151 (25)

151 (24)

137 (26)

135 (27)

AUC0–inf, ng·h/mLa

170 (26)

166 (29)

154 (23)

151 (21)

Cmax, ng/mL

45.0 (26)

45.6 (25)

25.0 (53)

22.4 (32)

tmax, h

0.75 (0.50–1.50)

0.75 (0.50–1.50)

2.00 (0.50–4.00)

2.00 (0.50–4.03)

t1/2, ha

10.5 (2.5)

9.5 (3.0)

10.0 (2.2)

10.2 (2.3)

Note: Geometric mean (arithmetic mean CV%) is reported only for AUCs and Cmax. Median (range) is presented for tmax. Arithmetic mean (SD) is presented for t1/2. a

The sample sizes for these parameters were N = 17 for the HS dapagliflozin 2.5 mg administered under fasting conditions and N =

25 for each of the other three treatments because some profiles did not have sufficiently well-defined Kel to include in the analysis. AUC0–inf, area under the plasma concentration–time curve from time 0 to infinity; AUC0–t, area under the plasma concentration–time curve from time 0 to the last quantifiable concentration; Cmax, maximum plasma concentration; CV, coefficient of variation; HS, heatstressed; Kel, terminal rate constant; NH, non–heat-stressed; SD, standard deviation; tmax, time to Cmax; t1/2, drug terminal half-life. 24

Table 4. Statistical analysis of plasma pharmacokinetic parameters of dapagliflozin 10-mg tablets. Parameter

Treatment

N

Geometric LS

Ratio (%) of geometric LS

mean

means (90% CI) (B/A or C/B)

AUC0–t, ng·h/mL

AUC0–inf, ng·h/mLa

Cmax, ng/mL

A: NH dapagliflozin 10 mg, fasted

28

637

B: HS dapagliflozin 10 mg, fasted

29

629

98.8 (95.9–101.8)

C: HS dapagliflozin 10 mg, fed

29

604

96.1 (93.2–99.0)

A: NH dapagliflozin 10 mg, fasted

28

663

B: HS dapagliflozin 10 mg, fasted

28

650

99.0 (96.0–102.1)

C: HS dapagliflozin 10 mg, fed

28

635

97.3 (94.3–100.4)

A: NH dapagliflozin 10 mg, fasted

28

172

B: HS dapagliflozin 10 mg, fasted

29

176

101.8 (92.3–112.2)

C: HS dapagliflozin 10 mg, fed

29

97

55.0 (49.9–60.6)

a

Some profiles did not have sufficiently well-defined Kel to include in the analysis for this parameter.

AUC0–inf, area under the plasma concentration–time curve from time 0 to infinity; AUC0–t, area under the plasma concentration–time curve from time 0 to the last quantifiable concentration; CI, confidence interval; Cmax, maximum plasma concentration; HS, heatstressed; Kel, terminal rate constant; LS, least squares; NH, non–heat-stressed.

25

Table 5. Statistical analysis of plasma pharmacokinetic parameters of dapagliflozin 2.5-mg tablets. Parameter

Treatment

N

Geometric LS

Comparison

mean AUC0–t, ng·h/mL

AUC0–inf, ng·h/mLa

Cmax, ng/mL

Ratio (%) of geometric LS means (90% CI)

D: NH dapagliflozin 2.5 mg, fasted

28

151





E: HS dapagliflozin 2.5 mg, fasted

26

150

E/D

99.0 (96.2–101.9)

F: NH dapagliflozin 2.5 mg, fed

27

137

F/D

90.3 (87.8–92.9)

G: HS dapagliflozin 2.5 mg, fed

28

135

G/E

90.3 (87.7–93.0)

D: NH dapagliflozin 2.5 mg, fasted

25

169





E: HS dapagliflozin 2.5 mg, fasted

17

165

E/D

97.6 (93.7–101.6)

F: NH dapagliflozin 2.5 mg, fed

25

156

F/D

92.5 (89.4–95.8)

G: HS dapagliflozin 2.5 mg, fed

25

155

G/E

94.0 (90.2–97.9)

D: NH dapagliflozin 2.5 mg, fasted

28

45





E: HS dapagliflozin 2.5 mg, fasted

26

45

E/D

99.8 (88.4–112.7)

F: NH dapagliflozin 2.5 mg, fed

27

25

F/D

55.6 (49.3–62.7)

G: HS dapagliflozin 2.5 mg, fed

28

22

G/E

49.9 (44.2–56.3)

26

a

Some profiles did not have sufficiently well-defined Kel to include in the analysis for this parameter.

AUC0–inf, area under the plasma concentration–time curve from time 0 to infinity; AUC0–t, area under the plasma concentration–time curve from time 0 to the last quantifiable concentration; CI, confidence interval; Cmax, maximum plasma concentration; HS, heatstressed; Kel, terminal rate constant; LS, least squares; NH, non–heat-stressed.

27

Table 6. Adverse event summaries. Study 1: Participants receiving dapagliflozin 10-mg tablets Parameter

Treatment A (N = 28)

B (N = 29)

C (N = 29)

2

1

11

Number of unique participants with ≥1 AE, n (%)

2 (7.1)

1 (3.4)

6 (20.7)

Diarrhea

1 (3.6)

1 (3.4)

0

Fatigue

0

0

2 (6.9)

Headache

0

0

2 (6.9)

Nausea

0

0

2 (6.9)

Feeling of body temperature change

0

0

1 (3.4)

Flatulence

0

0

1 (3.4)

Hypersensitivity

0

0

1 (3.4)

Pain

0

0

1 (3.4)

Retching

0

0

1 (3.4)

1 (3.6)

0

0

AEs, n

Vulvovaginal mycotic infection

Study 2: Participants receiving dapagliflozin 2.5-mg tablets Parameter

Treatment D (N = 28)

E (N = 26)

5

1

2

5

Number of unique participants with ≥1 AE, n (%)

4 (14.3)

1 (3.8)

2 (7.4)

4 (14.3)

Headache

1 (3.6)

0

0

2 (7.1)

Nausea

2 (7.1)

0

1 (3.7)

1 (3.6)

Conjunctival hyperemia

1 (3.6)

0

0

0

Dizziness

1 (3.6)

0

0

0

AEs, n

28

F (N = 27) G (N = 28)

Dry mouth

0

0

0

1 (3.6)

Fatigue

0

1 (3.8)

0

0

Gastroenteritis

0

0

1 (3.7)

0

Muscle spasm

0

0

0

1 (3.6)

Study 1: Treatment A, single oral dose of NH dapagliflozin 10 mg, fasted; Treatment B, single oral dose of HS dapagliflozin 10 mg, fasted; Treatment C, single dose of HS dapagliflozin 10 mg, fed. Study 2: Treatment D, single oral dose of NH dapagliflozin 2.5 mg, fasted; Treatment E, single oral dose of HS dapagliflozin 2.5 mg, fasted; Treatment F, single oral dose of NH dapagliflozin 2.5 mg, fed; Treatment G, single oral dose of HS dapagliflozin 2.5 mg, fed. No AEs leading to study discontinuation were reported in either study. AE, adverse event; HS, heat-stressed; NH, non–heat-stressed.

29

Figure legends Fig. 1. Study designs of (A) study 1 and (B) study 2. Washout = at least 4 days between doses. Study 1: Treatment A, single oral dose of NH dapagliflozin 10 mg, fasted; Treatment B, single oral dose of HS dapagliflozin 10 mg, fasted; Treatment C, single dose of HS dapagliflozin 10 mg, fed. Study 2: Treatment D, single oral dose of NH dapagliflozin 2.5 mg, fasted; Treatment E, single oral dose of HS dapagliflozin 2.5 mg, fasted; Treatment F, single oral dose of NH dapagliflozin 2.5 mg, fed; Treatment G, single oral dose of HS dapagliflozin 2.5 mg, fed. D, study discharge; HS, heat-stressed; NH, non–heat-stressed; R, randomization; S & E, screening and enrollment; W, washout.

Fig. 2. (A) Mean (+SD) plasma concentrations of dapagliflozin vs. time in study 1 and (B) in study 2 (linear scale). Study 1: Treatment A, single oral dose of NH dapagliflozin 10 mg, fasted; Treatment B, single oral dose of HS dapagliflozin 10 mg, fasted; Treatment C, single dose of HS dapagliflozin 10 mg, fed. Study 2: Treatment D, single oral dose of NH dapagliflozin 2.5 mg, fasted; Treatment E, single oral dose of HS dapagliflozin 2.5 mg, fasted; Treatment F, single oral dose of NH dapagliflozin 2.5 mg, fed; Treatment G, single oral dose of HS dapagliflozin 2.5 mg, fed. HS, heat-stressed; NH, non–heat-stressed; SD, standard deviation.

30