DMD Fast Forward. Published on November 8, 2017 as DOI: 10.1124/dmd.117.078006 This article has not been copyedited and formatted. The final version may differ from this version.

DMD # 078006 Simultaneous physiologically based pharmacokinetic (PBPK) modeling of parent and active metabolites to investigate complex CYP3A4 drug- potential: a case example of midostaurin

Helen Gu, Catherine Dutreix, Sam Rebello, Taoufik Ouatas, Lai Wang, Dung Yu Chun, Heidi J.

Einolf, Handan He

Downloaded from

Primary affiliations of all authors: HG, SR, LW, DYC, HJE, and HH: Novartis Pharmaceuticals

Corporation, East Hanover, New Jersey, USA. CD and TO: Basel, Switzerland. DYC: Insmed dmd.aspetjournals.org Inc., Bridgewater, New Jersey, USA.

Laboratory of origin: same as affiliations at ASPET Journals on September 30, 2021

DMD Fast Forward. Published on November 8, 2017 as DOI: 10.1124/dmd.117.078006 This article has not been copyedited and formatted. The final version may differ from this version.

RUNNING TITLE PAGE

Running title: PBPK modeling for midostaurin-CYP3A4 interaction

Corresponding author: Helen Gu, Novartis Pharmaceuticals Corporation, One Health Plaza, East

Hanover, NJ 07936 telephone: 1-862-778-5722 fax: 1-973-781-7579 Downloaded from e-mail: [email protected]

dmd.aspetjournals.org Text pages: 23

Tables: 5

Figures: 7 at ASPET Journals on September 30, 2021

References: 38

Word count:

Abstract: 249

Introduction: 748

Discussion: 1500

2

DMD Fast Forward. Published on November 8, 2017 as DOI: 10.1124/dmd.117.078006 This article has not been copyedited and formatted. The final version may differ from this version.

Abbreviations 4βHC, 4β-hydroxycholesterol ADME, absorption, distribution, metabolism, and excretion advSM, advanced systemic mastocytosis AGP, α1-acid glycoprotein AML, acute myeloid leukemia AUC, area under the curve bid, twice daily B/P, blood-to-plasma ratio Caco-2, continuous heterogeneous human epithelial colorectal adenocarcinoma cell line CL, clearance CL/F, apparent oral clearance

CLint, intrinsic clearance Downloaded from CLR, renal clearance Cmax, maximum concentration CYP, cytochrome P450 CYP3A4, cytochrome P450 3A4 DDI, drug-drug interaction dmd.aspetjournals.org EC50, concentration of inducer at half Emax Emax, maximal induction Fa, fraction of dose absorbed Fg, fraction of the dose that escapes presystemic intestinal first-pass elimination FLT3, fms-like tyrosine kinase 3 at ASPET Journals on September 30, 2021 fmCYP3A4, fraction metabolized by CYP3A4 fmCGP52421, fractions of total CGP52421 metabolism catalyzed by CYP3A4 fmCGP62221, fractions of total CGP62221 metabolism catalyzed by CYP3A4 fmother 3A4, fractions of total other metabolite metabolism catalyzed by CYP3A4 FMI, final market image fugut, fraction of unbound drug in the gut fumic, fraction of unbound drug in microsomes fup, fraction of unbound drug in plasma GM, geometric mean GMR, geometric mean ratio HLM, human liver microsome Ind50, half-maximum induction rate Indmax, maximum induction rate inf, infinity ITD, internal tandem duplication ka, absorption rate constant KI, concentration producing a half-maximal rate of activity reduction kin, zero-order rate constant for the formation of a physiological response variable kinact, maximal rate of activity reduction Kiu, unbound inhibition constant kout, first-order rate constant for the degradation of a physiological response variable LC-MS/MS, liquid chromatography–tandem mass spectrometry logPo:w, water partition coefficient MW, molecular weight 3

DMD Fast Forward. Published on November 8, 2017 as DOI: 10.1124/dmd.117.078006 This article has not been copyedited and formatted. The final version may differ from this version.

NA, not applicable PBPK, physiologically based pharmacokinetic Peff, man, effective permeability in man PK, pharmacokinetics pKa, acid dissociation constant Pop PK, population pharmaceutics Q, intercompartmental clearance qd, once daily Qgut, nominal flow through the gut SM, systemic mastocytosis TDI, time-dependent inhibitor TKD, tyrosine kinase domain Downloaded from tlag, lag time Tmax, time at which Cmax is reached Vsac, volume of distribution for the single adjustable compartment Vss, volume of distribution at steady state Vz/F, apparent volume of distribution dmd.aspetjournals.org

at ASPET Journals on September 30, 2021

4

DMD Fast Forward. Published on November 8, 2017 as DOI: 10.1124/dmd.117.078006 This article has not been copyedited and formatted. The final version may differ from this version.

ABSTRACT

Midostaurin (PKC412) is being investigated for the treatment of acute myeloid leukemia

(AML) and advanced systemic mastocytosis (advSM). It is extensively metabolized by

cytochrome P450 (CYP) 3A4 to form 2 major active metabolites, CGP52421 and CGP62221. In

vitro and clinical drug-drug interaction (DDI) studies indicated that midostaurin and its

metabolites are substrates, reversible and time-dependent inhibitors, and inducers of CYP3A4. A Downloaded from simultaneous pharmacokinetic model of parent and active metabolites was initially developed by

incorporating data from in vitro, preclinical, and clinical pharmacokinetic studies in healthy

volunteers and in patients with AML or advSM. The model reasonably predicted changes in dmd.aspetjournals.org midostaurin exposure after single-dose administration with (5.8-fold predicted vs

6.1-fold observed increase) and (90% predicted vs 94% observed reduction) as well as

changes in midazolam exposure (1.0 predicted vs 1.2 observed ratio) after daily dosing of at ASPET Journals on September 30, 2021 midostaurin for 4 days. The qualified model was then applied to predict the DDI effect with other CYP3A4 inhibitors or inducers and DDI potential with midazolam under steady-state conditions. The simulated midazolam AUC ratio of 0.54 and accompanying observed 1.9-fold increase in the CYP3A4 activity biomarker 4β-hydroxycholesterol indicated a weak-to-moderate

CYP3A4 induction by midostaurin and its metabolites at steady state in patients with advSM. In conclusion, a simultaneous parent-and-active-metabolite modeling approach allowed predictions under steady-state conditions that were not possible to achieve in healthy subjects. Furthermore,

endogenous biomarker data enabled evaluation of the net effect of midostaurin and its

metabolites on CYP3A4 activity at steady state and increased confidence in DDI predictions.

5

DMD Fast Forward. Published on November 8, 2017 as DOI: 10.1124/dmd.117.078006 This article has not been copyedited and formatted. The final version may differ from this version.

INTRODUCTION

Midostaurin is a potent kinase inhibitor of fms-like tyrosine kinase 3 (FLT3), KIT

(including D816V and Y mutants), platelet-derived growth factor receptor beta, vascular

endothelial growth factor receptor 2, fibroblast growth factor receptor, and protein kinase C.

FLT3 mutations occur in approximately 30% of patients with acute myeloid leukemia (AML).

Midostaurin is equally active against FLT3 with internal tandem duplications (ITDs) and with Downloaded from tyrosine kinase domain (TKD) mutations and has been shown to inhibit other kinases implicated in diseases, including KIT in advanced systemic mastocytosis (advSM) (Gotlib et al., 2005;

Weisberg et al., 2002; Barry et al., 2007; Andrejauskas-Buchdunger and Regenass, 1992; Fabbro dmd.aspetjournals.org et al., 2000; Stone et al., 2005; Propper et al., 2001). Midostaurin is in clinical development for

FLT3-mutated AML and advSM (Stone et al., 2005; Gallogly and Lazarus, 2016; Gotlib et al.,

2016; Fischer et al., 2010). Based primarily on the phase 3 RATIFY clinical study (Stone et al., at ASPET Journals on September 30, 2021

2015), the US Food and Drug Administration (FDA) granted approval to midostaurin in April

2017.

Midostaurin is rapidly and almost completely absorbed in humans after oral administration (Yin et al, 2008; Dutreix et al, 2013) and is highly distributed. Midostaurin is extensively metabolized by cytochrome P450 (CYP) 3A4 to form its 2 metabolites, CGP52421

(7-hydroxylation, epimer 2) and CGP62221 (O-demethylation) (Yin et al., 2008; Wang et al.,

2008). Elimination is primarily through fecal excretion, mostly as metabolites. CGP52421 and

CGP62221 have also been identified as being pharmacologically active against cells expressing

FLT3-ITDs and KIT D816V (Propper et al., 2001). Midostaurin (concentration of inducer at half

maximal induction of cytotoxicity response [EC50] 39 and 47 nM for FLT3-ITDs and KIT

D816V, respectively) and CGP62221 (EC50 30 and 70 nM for FLT3-ITDs and KIT D816V,

6

DMD Fast Forward. Published on November 8, 2017 as DOI: 10.1124/dmd.117.078006 This article has not been copyedited and formatted. The final version may differ from this version.

respectively) showed similar potency (Levis et al., 2006), whereas CGP52421 showed

approximately average 10-fold reduced potency (EC50 656 and 233 nM for FLT3-ITDs and KIT

D816V, respectively). Both metabolites were also found to be CYP3A4 substrates (Dutreix et al.

2013). Therefore, drug-drug interactions (DDIs) can result in exposure alterations of midostaurin and its metabolites when co-administered with drugs that inhibit or induce CYP3A4.

Furthermore, midostaurin and its 2 active metabolites were reversible and time-dependent inhibitors and inducers of CYP3A4 in vitro. These mixed CYP3A4 interactions can potentially Downloaded from

affect the exposure of victim drugs when co-administered with drugs that are sensitive CYP3A4

substrates (eg, midazolam). Due to these complex DDI mechanisms, midostaurin and its dmd.aspetjournals.org metabolites can act as CYP3A4 substrates, inhibitors, and inducers and also affect their own

metabolic clearance (FDA 2012; EMA 2012; Prueksaritanont et al., 2013; Fenneteau et al., 2010;

Fahmi et al., 2009; Garg et al., 2012; Yeo et al., 2011; Reitman et al., 2011) or that of other at ASPET Journals on September 30, 2021

drugs.

In the initial phase of development, DDI studies were conducted in healthy volunteers to

address the clinical relevance of CYP3A4-related DDIs, with midostaurin as either a victim of a

ketoconazole or rifampicin interaction or a perpetrator of midazolam (Dutreix et al., 2013;

Dutreix et al., 2014). However, these early clinical studies were conducted mostly using

relatively short dosing periods. The terminal half-lives of midostaurin, CGP62221, and

CGP52421 in plasma are 19.6, 32.2, and 482 hours, respectively. Due to the complex DDI mechanisms of midostaurin and its metabolites, it is important to evaluate potential DDIs under steady-state conditions to support clinical recommendations and potential product label language. Because such DDI studies are not feasible in healthy subjects, we aimed to use a multipronged approach to investigate the net effect of steady-state midostaurin on CYP3A4

7

DMD Fast Forward. Published on November 8, 2017 as DOI: 10.1124/dmd.117.078006 This article has not been copyedited and formatted. The final version may differ from this version.

activity. To achieve this goal, we (1) built a physiologically based pharmacokinetic (PBPK)

model to describe the pharmacokinetics (PK) of midostaurin and its 2 active metabolites using a

top-down approach leveraging prior clinical PK data in both healthy subjects and patients; (2) verified the model’s performance in predicting clinical PK profiles of midostaurin and its metabolites, particularly in simulating clinically observed DDIs under single-dose conditions; and (3) used the model to predict CYP3A4-related DDIs with midostaurin at steady state.

Furthermore, the utility of the endogenous plasma biomarker 4β-hydroxycholesterol (4βHC) as a Downloaded from tool for assessing CYP3A4 activity was explored to address the uncertainty in evaluating the net effect of CYP3A4 induction or inhibition of new molecular entities that have mixed inhibition or dmd.aspetjournals.org induction DDI mechanisms in vitro. The final model was also used to predict DDI potential with other CYP3A4 substrates, inhibitors, and inducers; the model limitations are discussed. The prediction results from our case example provide a basis for optimal clinical study at ASPET Journals on September 30, 2021 recommendations and potential label language.

MATERIALS AND METHODS

PBPK modeling was accomplished following these general steps: (1) the model was

initially developed using physiochemical data; absorption, distribution, metabolism, and excretion (ADME) data; and in vitro DDI study data and optimized to fit the clinical PK data of the parent and its 2 metabolites; (2) the model was verified by comparing simulated data with data from clinical DDI studies; and (3) the model was applied to predict potential DDI effects of

CYP3A4 inhibitors or inducers on midostaurin and its metabolites as well as effects on midazolam under steady-state conditions. Finally, the results from plasma 4βHC levels (fold

8

DMD Fast Forward. Published on November 8, 2017 as DOI: 10.1124/dmd.117.078006 This article has not been copyedited and formatted. The final version may differ from this version.

change relative to baseline) were used to confirm the net effect of time-dependent

inhibition/induction of CYP3A4 by midostaurin and its metabolites.

PBPK Model Development in Healthy Subjects

A PBPK model was built using Simcyp Simulator, version 15, release 1 (Certara, LP,

Princeton, NJ) for midostaurin and its 2 metabolites, CGP52421 and CGP62221, using in vitro

ADME and in vivo PK data (Table 1). The key input parameters are described here.

Physiochemical Properties and Plasma Binding Downloaded from

The molecular weights of midostaurin, CGP52421, and CGP62221 are 570.6, 586.6, and

556.6 g/mol, respectively, and the water partition coefficient (logP octanol:water) ratios used dmd.aspetjournals.org were 5.5, 4.8, and 4.7 (calculated logP values), respectively. The compound type was entered as

a monoprotic base, with a pKa value of 11.2 for midostaurin and CGP62221 and a pKa value of

10.8 for CGP52421 (predicted using ADMET Predictor 6.5, Simulations Plus, Lancaster, CA). at ASPET Journals on September 30, 2021

The blood-to-plasma ratios for midostaurin, CGP62221, and CGP52421 were entered as 0.55

based on the internal experimental values.

The fractions bound to plasma for all 3 components were very high (> 99%). For these

very highly bound compounds, an equilibrium gel filtration method was used (He et al., 2017;

Weiss et al., 2014). Midostaurin was mainly bound to human α1-acid glycoprotein (AGP)

according to an internal study. Therefore, AGP was selected as a plasma-binding component in

the model.

Absorption

A first-order model for midostaurin absorption from the gut lumen was chosen. Several

key input parameters were applied. First, the fraction of dose absorbed (Fa) was entered as 0.85,

primarily to optimize maximum concentration (Cmax), based on the observed data from a single

9

DMD Fast Forward. Published on November 8, 2017 as DOI: 10.1124/dmd.117.078006 This article has not been copyedited and formatted. The final version may differ from this version.

oral midostaurin 50-mg dose in a prior clinical study. This Fa value was consistent with the

moderate to high absorption observed in preclinical species and humans (He et al., 2017). The absorption rate constant and lag time were user defined as 1.5 1/h and 0.3 hours, respectively, to optimize Cmax and Tmax that were observed in several clinical trials (after a single dose). Second,

the fraction of unbound drug in the gut (fugut) was predicted to be 0.3, and, accordingly, by

Simcyp, the output fraction of the dose that escapes presystemic intestinal first-pass elimination Downloaded from (Fg) value was 0.15, a value derived from the Simcyp-predicted fugut value. This predicted human

Fg was consistent with the observed rat Fg based on calculations from data obtained in a rat in

vivo study (data not shown). Finally, the nominal flow through the gut value captures the fact dmd.aspetjournals.org that a higher permeability past the enzyme will decrease first-pass exposure to the enzyme, as

will a greater blood flow carrying drug away from the enterocytes. The value was predicted to be

5.23 L/h with Simcyp, using the apparent permeability data from an internal continuous at ASPET Journals on September 30, 2021

heterogeneous human epithelial colorectal adenocarcinoma cell line (Caco-2) study.

Distribution

The minimal PBPK model in Simcyp was used with a single adjusting compartment for

all 3 components. The volume of distribution at steady state (Vss) was estimated to be 1 L/kg, with parameters for a Q (intercompartmental clearance) of 3 L/h and a volume of distribution for the single adjustable compartment (Vsac) of 0.82 L/kg. The estimated midostaurin oral Vss was

close to the observed geometric mean (GM) or median apparent volume of distribution values, which ranged from 1.0 to 1.3 L/kg. For CGP52421, Vss was estimated to be 1.8 L/kg, with a Q of

10 L/h and a Vsac of 1.3 L/kg. For CGP62221, Vss was estimated to be 1.3 L/kg, with a Q of 2

L/h and a Vsac of 1.1 L/kg. All values were estimated based on optimization to best fit to PK data

from clinical trials (Supplemental Table 9).

10

DMD Fast Forward. Published on November 8, 2017 as DOI: 10.1124/dmd.117.078006 This article has not been copyedited and formatted. The final version may differ from this version.

Elimination

The retrograde model was used to calculate in vitro intrinsic clearance (CLint) values of

the relevant metabolizing enzymes from intravenous or apparent oral clearance (CL/F). In brief,

the CL/F of 2.4 L/h was estimated to optimize the plasma concentration-time profiles of

midostaurin observed in the clinical trials used for model development. This estimated oral

clearance value was consistent with the observed median or GM CL/F values, which ranged Downloaded from from 2.1 to 3.8 L/h. The fraction metabolized by CYP3A4 (fmCYP3A) was set to a value of 1 for midostaurin based on human in vitro findings and the human ADME study. As such, CYP3A4

CLint was calculated to be ≈25.2 µL/min/mg/pmol of CYP3A4 by the retrograde model for dmd.aspetjournals.org

enzyme kinetics in Simcyp. In a second step, the CYP3A4 CLint was divided between the

involved elimination pathways (ie, CGP52421, CGP62221, and other CYP3A4-mediated pathway[s]). The fraction of CYP3A4 for the metabolism (formation of CGP52421 and at ASPET Journals on September 30, 2021

CGP62221)/elimination of midostaurin was estimated. The estimations are based mainly on an

enzyme phenotyping study using human liver microsomes (HLMs) and CYP-selective chemical

inhibitors; the estimations showed that CYP3A4 contributed to most of the hepatic oxidative

microsomal metabolism of midostaurin. Furthermore, CGP52421 and CGP62221 are also

metabolized mainly by CYP3A4. The human ADME study (He et al., 2017) showed that

midostaurin undergoes extensive metabolism and is predominantly excreted through feces,

mostly as metabolites. The fractions of CYP3A4 for the metabolism/elimination of midostaurin

in human fecal excreta were estimated to be 37%, 37%, and 26% for CGP52421, CGP62221,

and other minor metabolites, respectively. Consequently, the values of CLint in µL/min/pmol of

CYP3A4 were assigned to the following CYP3A4 pathways: CGP52421, 9.3; CGP62221, 9.3; other CYP3A4, 6.6 (µL/min/pmol of CYP3A4).

11

DMD Fast Forward. Published on November 8, 2017 as DOI: 10.1124/dmd.117.078006 This article has not been copyedited and formatted. The final version may differ from this version.

For the oral CL/F, the values were estimated to be 0.4 and 0.27 L/h for CGP52421 and

CGP62221, respectively, based on optimization to best fit to the plasma concentration-time

profiles of these metabolites from clinical trials. Furthermore, it was assumed that approximately

90% of CGP52421 and CGP62221 clearance was mediated by CYP3A4, as they were detected at

low levels in human excreta. The CYP3A4 CLint values derived from the CL/F were predicted by the retrograde model to be 3.18 µL/min/pmol of CYP3A4 and 1.19 µL/min/pmol of CYP3A4 for

CGP52421 and CGP62221, respectively. Downloaded from

Data from the human ADME study indicated that midostaurin in urine was not detected, as shown in a high-performance liquid chromatography profile of radioactive components in dmd.aspetjournals.org pooled urine (0-72 h). Therefore, the renal CL was assumed to be negligible and set to 0 L/h.

Interaction

The potential of midostaurin, CGP52421, and CGP62221 to inhibit human CYP3A4/5 at ASPET Journals on September 30, 2021 enzyme activity and act as CYP3A4/5 time-dependent inhibitors (TDIs) was assessed using pooled HLMs (Supplemental Data). The potential for midostaurin, CGP52421, and CGP62221 to act as inducers of CYP3A4/5 enzymes was also evaluated in primary human hepatocytes of 3 individual donors using both mRNA quantification (real-time polymerase chain reaction) and

CYP3A4/5 activity measurements (Supplemental Data). All of the in vitro enzyme activity was determined using liquid chromatography–tandem mass spectrometry (LC-MS/MS) of selective

CYP3A4/5 probe substrate metabolism (Supplemental Data). The input parameters used for induction of CYP3A4, TDI, and reversible inhibition of CYP3A4/5 are summarized in Table 1.

Model Assumptions

Several assumptions were made for projection of human PK parameters. First, human Fa

was set to 0.85 (see Absorption). Next, the fractions of total metabolism catalyzed by CYP3A4

12

DMD Fast Forward. Published on November 8, 2017 as DOI: 10.1124/dmd.117.078006 This article has not been copyedited and formatted. The final version may differ from this version.

in humans were estimated to be fmCGP52421 ≈0.37, fmCGP62221 ≈0.37, and fmother 3A4 ≈0.26, with the

assumptions that metabolite epimer structures were similar and that sequentially formed

metabolites were equally contributed by primary and secondary metabolism. The CYP3A4/5

TDI KI (total) was used. The CYP3A4 mRNA induction EC50 values of midostaurin, CGP52421,

and CGP62221 were entered and optimized in the models (ie, 20-fold more potent than the

experimental value). These modifications (ie, lower EC50 and fixed maximal induction [Emax])

were judged to be appropriate based on the very high plasma protein binding of all 3 components Downloaded from

observed (> 99%); lower intracellular free concentrations in hepatocytes would be expected,

which could reflect the amount of drug available for binding to the . The dmd.aspetjournals.org model was developed and verified using data from clinical studies in healthy subjects (further

details follow). The disease- and/or age-related physiological changes affecting drug ADME

properties, including enzyme activity and function, were not considered. Finally, the transporter- at ASPET Journals on September 30, 2021

related drug interaction property was not included in the model.

Population

The simulations were performed using a simulated healthy volunteer population built in

the Simcyp simulator, with individuals aged 20 to 55 years; the proportion of female subjects

was set to 50%. The population size was 100, with 10 trials of 10 subjects per trial.

Simulation Trials

The simulated trials were run in a fasted state. The simulated trials for plasma PK of

midostaurin and its 2 metabolites after single and multiple twice-daily (bid) doses are presented

as follows. First, observed and predicted PK parameters and plasma concentration-time profiles

of midostaurin, CGP52421, and CGP62221 following a single dose of midostaurin with a final

market image (FMI) formulation of 50 mg in the simulated subjects were compared with those in

13

DMD Fast Forward. Published on November 8, 2017 as DOI: 10.1124/dmd.117.078006 This article has not been copyedited and formatted. The final version may differ from this version.

healthy volunteers in clinical studies. Control arms included food effect, relative bioavailability,

DDIs with ketoconazole, and DDIs with rifampicin. Second, observed and predicted PK

parameters and plasma concentration-time profiles of midostaurin, CGP52421, and CGP62221

following midostaurin (FMI) 75 mg bid on days 1 and 2 and 75 mg once daily (qd) on day 3 in

the simulated subjects were compared with those in healthy volunteers in a cardiac intervals

investigation. Third, observed and predicted PK parameters and plasma concentration-time

profiles of midostaurin, CGP52421, and CGP62221 following midostaurin (FMI) 50 mg bid on Downloaded from

days 1 to 6 and a single dose on day 7 in the simulated subjects were compared with those in

healthy subjects with normal hepatic function in a PK study in subjects with impaired hepatic dmd.aspetjournals.org function.

PBPK Model Performance

The performance of the PBPK model was verified by comparing the simulated data with at ASPET Journals on September 30, 2021 clinically observed DDIs. Input parameters for the PBPK model are in Supplemental Tables 10-

15. The ketoconazole, rifampicin, and midazolam models available in the Simcyp compound library were used in the simulations. The simulated trials for plasma PK of midostaurin and its 2 metabolites in the presence of CYP3A4/5 inhibitors, a CYP3A4 inducer, or sensitive CYP3A4

substrates are shown in Table 2.

PBPK Model Development in Patients With AML and AdvSM

A PBPK model was also built to simulate the PK of midostaurin and its metabolites in

patients with AML and advSM using a top-down approach. The model was mainly adopted from

the previous Simcyp model in healthy subjects but with modifications. The assumption of these 2

patient models was that the protein binding, fmCYP3A4, and in vitro DDI parameters of

midostaurin and its metabolites were the same in patients and healthy subjects. The parameters

14

DMD Fast Forward. Published on November 8, 2017 as DOI: 10.1124/dmd.117.078006 This article has not been copyedited and formatted. The final version may differ from this version.

were updated based on the parameter estimated in the population (Pop) PK of midostaurin in

patients with AML and advSM. These values were entered as described in the model for healthy

subjects, with the exception of the following changes (Table 1).

For patients with AML, parameters of distribution and elimination for midostaurin and

parameters of elimination for CGP52421 were updated in the model for healthy subjects. Vss of

midostaurin was changed from 1 to 0.742 L/kg, with changes in parameters for Q (3 to 2.889 Downloaded from

L/h) and Vsac (0.82 to 0.623 L/kg). CLint of midostaurin was changed from ≈25.2 to 10.6

µL/min/mg/pmol of isoform. Accordingly, the midostaurin value of CLint (in μL/min/pmol of

CYP3A4) was assigned to the following CYP3A4 pathways: CGP52421 (37%) = 3.9; dmd.aspetjournals.org

CGP62221 (37%) = 3.9; other (26%) = 2.8. For CGP52421, CL/F and CLint were changed from

0.4 to 0.07 L/h and from 3.18 to 0.557 μL/min/mg/pmol of CYP3A4, respectively, with

additional HLM clearance of 6.48 μL/min/mg of protein. Comparison of PBPK- and Pop PK– at ASPET Journals on September 30, 2021

simulated data can be found in Supplemental Table 5, indicating that PBPK-simulated AUC and

Cmax values were within ≤ 2-fold of the values in the Pop PK data analysis.

For patients with advSM, parameters of absorption, distribution, and elimination for

midostaurin were updated in the model for healthy subjects, so that the same Vss and CLint of midostaurin used in the model for patients with AML were used in the model for patients with advSM. In addition, midostaurin Fa was changed from 0.85 to 0.65, and the absorption rate

constant was changed from 1.5 to 0.683 L/h. These values were updated based on optimization to

best fit to population PK data in patients with advSM. Comparison of PBPK- and Pop PK–

simulated data can be found in Supplemental Table 6, indicating that PBPK-simulated AUC and

Cmax values were within ≤ 2-fold of the values in the Pop PK data analysis.

15

DMD Fast Forward. Published on November 8, 2017 as DOI: 10.1124/dmd.117.078006 This article has not been copyedited and formatted. The final version may differ from this version.

PBPK Model Application

Following model verifications using data from clinical PK studies as well as clinical

DDIs with ketoconazole, rifampicin, and midazolam in healthy subjects, the model was used to simulate hypothetical DDI scenarios at steady-state levels in healthy subjects and in patients with

AML and advSM. These scenarios included (1) predicting the effect of moderate and strong

CYP3A4 inhibitors and inducers on the exposure of midostaurin and its metabolites and (2) predicting the effect of midostaurin on the exposure of midazolam. Downloaded from

Simulation of the Effects of Moderate and Strong CYP3A4 Inhibitors and Inducers

The potential effects of CYP3A4 inhibitors fluconazole (moderate), itraconazole (strong), dmd.aspetjournals.org and ketoconazole (strong) and those of inducers (moderate) and rifampicin (strong) on the PK of midostaurin and its metabolites were simulated. The Simcyp default PBPK models for

efavirenz, fluconazole, itraconazole (fed capsule) with OH-itraconazole, ketoconazole, and at ASPET Journals on September 30, 2021 rifampicin were used in these simulations. Next, midostaurin 50 mg bid was administered for 21 days; efavirenz 600 mg qd, fluconazole 200 mg qd, itraconazole 100 mg bid, ketoconazole 400 mg qd, or rifampicin 600 mg qd was co-administered with midostaurin for 7 days, starting on day 22. The effects of inhibitors and inducers on the PK of midostaurin were assessed after the last midostaurin dose on day 28.

Simulation of the Effects of Midostaurin and Its Metabolites on Midazolam

Due to a technical limitation in the current version of Simcyp, the model does not have the ability to enter midostaurin with 2 metabolites as perpetrators. Therefore, an alternative approach was used in which the simulations were performed using the perpetrator as the model’s substrate and the victim as the model’s inhibitor. Briefly, 2 simulations were conducted with midostaurin and its 2 metabolites in the substrate position and midazolam in the inhibitor

16

DMD Fast Forward. Published on November 8, 2017 as DOI: 10.1124/dmd.117.078006 This article has not been copyedited and formatted. The final version may differ from this version.

position. In the first simulation, the DDI properties of midostaurin and its metabolite CYP3A

were turned off, representing the control PK of midazolam (in the absence of interaction). The

second simulation was performed in the presence of midostaurin by including all the DDI

properties, representing the interaction between midazolam as a perpetrator on midostaurin and

its metabolites as victims. The exposure parameters [eg, area under the curve (AUC) and Cmax] of

midazolam from the first simulation were compared with those of the second simulation to

determine the effect of midostaurin and its metabolites on midazolam. The dose regimen was Downloaded from

midazolam 4 mg on day 28, with midostaurin 50 mg bid on days 1 to 28.

Plasma 4βHC Measurement dmd.aspetjournals.org

Study Design

Healthy adult volunteers aged 18 to 55 years were randomized to receive either

rifampicin 600 mg (positive control) or placebo (negative control) from day 1 to 14. All at ASPET Journals on September 30, 2021

individuals received midostaurin 50 mg on day 9. This unique, single administration was deemed not to interfere with the rifampicin-placebo effect. The positive control arm (n = 20) and negative

control arm (n = 20) served as references for the progression of 4βHC levels. In a separate study, data from patients with advSM enrolled in an open-label, phase 2 study who received midostaurin 100 mg bid for 28 days (n = 10) were used to determine the 4βHC concentration profile. The study was conducted in accordance with the Declaration of Helsinki and approved by all relevant institutional review boards and ethics committees. The study design is summarized in Table 3.

Sample Collection and Analysis

To evaluate the plasma concentration of 4βHC, peripheral blood samples were collected on days 1 (baseline), 9 (before receiving midostaurin), 11, and 15 in the positive and negative

17

DMD Fast Forward. Published on November 8, 2017 as DOI: 10.1124/dmd.117.078006 This article has not been copyedited and formatted. The final version may differ from this version.

control arms (healthy participants in a clinical DDI study with rifampicin) and on days 1, 3, 8,

15, 22, and 28 of cycle 1 in the patient study. The plasma concentration of 4βHC was measured

as described (Diczfalusy et al., 2011), using validated LC-MS/MS with a lower limit of

quantification of 3 ng/mL.

Statistical Analyses

Statistical analyses were descriptive only; no formal statistical tests were performed.

Only subjects with an available full 4βHC profile were included in the analysis. For each subject, Downloaded from values at each time point and percent change from baseline were considered and median values were computed (data not shown); the mean and GM of the individual value from baseline were dmd.aspetjournals.org also calculated. Results were plotted to visually represent the data and support the interpretations.

RESULTS at ASPET Journals on September 30, 2021

Simulated and Observed PK Following Single and Multiple Doses of Midostaurin in

Healthy Subjects

A Simcyp model was established to predict the PK of midostaurin, CGP52421, and

CGP62221 after single (50 mg) and multiple (50 or 75 mg) oral doses in healthy subjects.

Simulated PK profiles were compared with data from several clinical trials in which single-agent

PK was available. Overall, the PK of midostaurin, CGP52421, and CGP62221 was predicted

reasonably well. The simulated mean plasma concentration-time profiles with observed data

overlaid are shown in Figure 1. The representative tabulated simulated and actual PK parameters

for midostaurin, CGP52421, and CGP62221 following a single dose of 50 mg and 50 mg BID of

midostaurin can be found in Supplemental Tables 7 and 8.

18

DMD Fast Forward. Published on November 8, 2017 as DOI: 10.1124/dmd.117.078006 This article has not been copyedited and formatted. The final version may differ from this version.

Simulated and Observed DDIs With CYP3A4 Inhibitor Ketoconazole and Inducer

Rifampicin in Healthy Subjects

The clinical DDI trial of midostaurin 50 mg with ketoconazole 400 mg qd or rifampicin

600 mg qd was simulated according to the clinical trial design in healthy subjects. Simulated profiles for the interaction of midostaurin and ketoconazole or midostaurin and rifampicin were then compared with observed data, as shown in Figure 2. Downloaded from The predicted and observed midostaurin AUC GM ratios (GMRs) were 5.8 (AUCinf, 6.3)

and 6.1 (AUCinf, 10.4), respectively, and the predicted and observed Cmax GMRs were 2.1 and

1.8, respectively, when midostaurin was co-administered with ketoconazole. For CGP52421, the dmd.aspetjournals.org

predicted and observed GMRs were 0.6 and 1.2 for AUC and 0.4 and 0.5 for Cmax, respectively.

For CGP62221, the predicted and observed GMRs were 0.6 and 1.0 for AUC and 0.3 and 0.6 for

Cmax, respectively. When midostaurin was co-administered with rifampicin, the predicted vs at ASPET Journals on September 30, 2021

observed decrease in GM AUC and Cmax for midostaurin was 90% vs 94% and 79% vs 73%,

respectively. The predicted vs observed decrease in GM AUC for CGP52421 was 65% vs 59%,

respectively, and for CGP622221, 55% vs 92%, respectively. The corresponding predicted vs observed decrease in GM Cmax for CGP52421 and CGP62221 was 9% vs 35% and 37% vs no change, respectively. Overall, the model predicted the exposure change in midostaurin reasonably well when it was co-administered with ketoconazole or rifampicin. However, for its metabolites, there was a trend toward underprediction of exposure change compared with observed data (Table 4).

19

DMD Fast Forward. Published on November 8, 2017 as DOI: 10.1124/dmd.117.078006 This article has not been copyedited and formatted. The final version may differ from this version.

Simulated DDIs With Moderate and Strong CYP3A4 Inhibitors and Inducers at

Steady State in Healthy Subjects and Patients With AML and AdvSM

The potential effects of CYP3A4 inhibitors fluconazole (moderate), itraconazole (strong),

and ketoconazole (strong) as well as those of CYP3A4 inducers efavirenz (moderate) and

rifampicin (strong) on midostaurin exposure at steady-state levels (50 mg bid for healthy subjects

and patients with AML and 100 mg bid for patients with advSM for 28 days) were simulated Downloaded from both in healthy subjects and in patients with AML and advSM.

GMRs of AUC0-tau for itraconazole were predicted to be 2.5-, 1.3-, and 1.3-fold for

midostaurin, CGP52421, and CGP62221, respectively, using the model for healthy subjects. dmd.aspetjournals.org

GMR values of AUC0-tau for ketoconazole DDIs were predicted to be 5.4-, 1.6-, and 1.7-fold for

midostaurin, CGP52421, and CGP62221, respectively. The results indicated that the impact of

DDIs after multiple doses of midostaurin (to steady state) with ketoconazole was similar to the at ASPET Journals on September 30, 2021

impact of DDIs after a single midostaurin dose (Table 4). GMR values of AUC0-tau for midostaurin with fluconazole were predicted to be 2.7-, 1.6-, and 1.5-fold for midostaurin,

CGP52421, and CGP62221, respectively. GMR values of AUC0-tau for rifampicin DDIs were

predicted to decrease by 43%, 30%, and 25% for midostaurin, CGP52421, and CGP62221,

respectively. The impact of DDIs after multiple doses of midostaurin (to steady state) with

rifampicin appears to be less than the impact of DDIs after a single dose of midostaurin (Table

4). With co-administration of efavirenz, GMR values of AUC0-tau were predicted to decrease by

8%, 5%, and 4% for midostaurin, CGP52421, and CGP62221, respectively, suggesting a

minimal impact on exposure to midostaurin and its metabolites. The overall prediction results are

summarized in the forest plots shown in Figure 3.

20

DMD Fast Forward. Published on November 8, 2017 as DOI: 10.1124/dmd.117.078006 This article has not been copyedited and formatted. The final version may differ from this version.

GMR values of AUC0-tau for itraconazole for AML and advSM were predicted to be 2.2-

and 2.0-fold, 1.0- and 1.2-fold, and 1.2-fold for midostaurin, CGP52421, and CGP62221,

respectively, applying the models for patients with AML and advSM. GMR values of AUC0-tau

for ketoconazole DDIs for AML and advSM were predicted to be 4.2- and 4.4-fold, 1.1- and 1.5-

fold, and 1.5- and 1.6-fold for midostaurin, CGP52421, and CGP62221, respectively. GMR

values of AUC0-tau for midostaurin with fluconazole for AML and advSM were predicted to be

2.5- and 2.6-fold, 1.2- and 1.6-fold, and 1.5- and 1.6-fold for midostaurin, CGP52421, and Downloaded from

CGP62221, respectively. GMR values of AUC0-tau for rifampicin DDIs for AML and advSM were predicted to decrease by 27% and 34%%, 13% and 24%, and 21% and 20% for dmd.aspetjournals.org midostaurin, CGP52421, and CGP62221, respectively. With co-administration of efavirenz,

GMR values of AUC0-tau were predicted to decrease by 5% and 3%, 1% and 2%, and 3% and 2%

for midostaurin, CGP52421, and CGP62221, respectively. The overall prediction results are at ASPET Journals on September 30, 2021

summarized in the forest plots shown in Figure 3.

Simulated Hepatic CYP3A4 Dynamics After Treatment With CYP3A4 Inhibitors

and Inducers

From a victim DDI perspective, the model reasonably predicted the observed changes in

midostaurin exposure with ketoconazole and rifampicin. The qualified model for healthy subjects

was then applied to predict the DDI effect with ketoconazole or rifampicin at steady-state levels.

In the presence of a strong CYP3A4 inducer (rifampicin 600 mg qd), the ratios (relative to the control arm) of maximal increased hepatic CYP3A4 activity were 4.9 and 1.9 after a single dose and bid dosing of midostaurin (steady state), respectively (Figure 4A). These ratios are consistent with predicted reductions of midostaurin exposure by 90% (single dose) and 43% (steady state) due to the change in CYP3A4 enzyme activity. Conversely, in the presence of a strong CYP3A4

21

DMD Fast Forward. Published on November 8, 2017 as DOI: 10.1124/dmd.117.078006 This article has not been copyedited and formatted. The final version may differ from this version.

inhibitor (ketoconazole 400 mg qd), the ratios (relative to the control arm) of maximal increased

hepatic CYP3A4 activity were 1.08 and 1.02 after a single dose and bid dosing of midostaurin

(steady state), respectively (Figure 4B). These ratios are consistent with predicted similar

increases of midostaurin exposure by 5.8-fold (single dose) and 5.4-fold (steady state) due to the

similar change in CYP3A4 enzyme activity.

Simulated and Observed DDIs With CYP3A4 Substrate Midazolam

The clinical combination trial for a single dose of midazolam (4 mg) with a single dose of Downloaded from midostaurin (100 mg on day 1, followed by 50 mg bid on days 2 to 4) was simulated according to the clinical trial design. The effect of midostaurin on midazolam AUC0-inf was predicted to be dmd.aspetjournals.org

1.2-fold (observed, 1.0-fold) and 0.74-fold (observed, 0.95-fold) on days 1 and 6, respectively.

The corresponding change in midazolam Cmax was predicted to be 1.2-fold (observed, 0.8-fold)

and 1-fold (observed, 0.9-fold), respectively. These results indicated that no CYP3A4 induction at ASPET Journals on September 30, 2021 effect could be detected in a relatively short study duration. The study duration was short due to safety concerns for healthy subjects. The observed and simulated plasma concentration-time profiles are presented in Figure 5.

Simulated DDIs With CYP3A4 Substrate Midazolam at Steady State in Healthy

Subjects and Patients With AML and AdvSM

Simcyp Simulation

The potential interaction of midostaurin with the sensitive CYP3A4 substrate midazolam

under steady-state midostaurin dosing conditions was simulated in both healthy subjects and

patients with AML and advSM. With co-administration of midostaurin (50 mg bid in healthy

subjects) to steady state (ie, 28 days), midazolam AUCinf and Cmax ratios (relative to dose alone)

were predicted to be 0.59 (decreased by 41%) and 0.78 (decreased by 22%), respectively (Figure

22

DMD Fast Forward. Published on November 8, 2017 as DOI: 10.1124/dmd.117.078006 This article has not been copyedited and formatted. The final version may differ from this version.

6). The net decrease in midazolam exposure (AUCinf) in the presence of midostaurin (50 or 100

mg bid in AML and advSM, respectively) relative to dose alone was predicted to be 0.60 and

0.54, respectively. Overall, this would be indicative of an induction effect of midostaurin and its metabolites at steady state.

Assessment of CYP3A4 Activity Using Plasma Biomarker 4βHC Levels

At baseline, the GM 4βHC levels were similar between the negative control arm, positive

control arm, and midostaurin arm despite the absence of global randomization (Table 5). Downloaded from

In the negative control arm (subjects receiving placebo), 4βHC levels remained stable on

days 9, 11, and 15. No time dependency in levels was observed. In contrast, 4βHC levels notably dmd.aspetjournals.org increased over time in the positive control arm (subjects treated with rifampicin) on days 9, 11,

and 15.

In the midostaurin arm (100 mg bid), 4βHC levels moderately increased (≈ 1.8-fold) until at ASPET Journals on September 30, 2021

day 15, at which point they had approximately doubled relative to baseline. The 4βHC levels

remained constant from day 15 to PK steady state of midostaurin (day 28) (Figure 7). GM 4βHC levels on days 3, 8, 9, 11, 15, 22, and 28 are presented (Table 5). A net increase in plasma 4βHC was observed with midostaurin (100 mg bid) under steady-state conditions, also suggesting an induction effect of midostaurin and its metabolites at steady state. In the rifampicin arm (positive control), the 4βHC levels increased 3.2-, 3.5-, and 4.3-fold on days 9, 11, and 15, respectively.

DISCUSSION

In the current study, we present a case example using a PBPK modeling approach to predict the DDIs associated with a mixed net effect (TDI and induction) on CYP3A4 for the

parent compound as well as its 2 major circulating metabolites in healthy subjects and in patients

with AML and advSM.

23

DMD Fast Forward. Published on November 8, 2017 as DOI: 10.1124/dmd.117.078006 This article has not been copyedited and formatted. The final version may differ from this version.

In our modeling approach, in vitro study–determined parameters (Supplemental Data) were initially used. However, the simulated concentration-time profiles after multiple dosing showed net auto-inhibition (data not shown) rather than observed auto-induction. Subsequently,

EC50 values were optimized (20-fold more potent for all 3 components) based on fitting of

single- and multiple-dosing PK profiles from clinical PK and DDI trials. The under-prediction in

induction magnitude using in vitro data could be due to other mechanisms involved in the

induction of CYP3A4 mRNA. For example, CYP3A4 could be induced through P53 protein Downloaded from

activation and may be relevant for patients with cancer (Goldstein et al., 2013; Harmsen et al.,

2008; Elias et al., 2013); tyrosine kinase inhibitors may potentially induce CYP3A4 mRNA dmd.aspetjournals.org involving CAR1 and P53 (Lin et al., 2016). Because the induction was ultimately modeled using a “top-down” approach for midostaurin and its metabolites, any other potential mechanisms of

CYP3A4 regulation would be captured in the model. Although a more extensive data set was at ASPET Journals on September 30, 2021 required, this model is considered to be mechanistic because it allows for incorporation of changes in clearance of midostaurin as a result of its auto-inhibition/induction after multiple dosing. By using the middle-out approach (combining top-down and bottom-up), the model could reasonably capture the acute effects of auto-inhibition and subsequent auto-induction, as predicted by increased exposure of midostaurin on day 7 relative to day 1 followed by a decreasing trend in exposure on day 28. The half-life and time to steady state were taken into account, and a standard dosing regimen (50 mg bid for healthy subjects and patients with AML and 100 mg bid for patients with advSM for 28 days) was consistently used to closely mimic a potential clinical, steady-state dosing regimen. The model was then used to simulate the PK profiles of midostaurin and its metabolites at steady state and predict the subsequent effects on midazolam exposure.

24

DMD Fast Forward. Published on November 8, 2017 as DOI: 10.1124/dmd.117.078006 This article has not been copyedited and formatted. The final version may differ from this version.

Per regulatory guidance (FDA, 2012; EMA, 2012), the simulated midazolam AUC ratio

is consistent with the classification of midostaurin as a weak CYP3A4 inducer (20%-50% decrease in AUC). The simulated net effect of midostaurin and its metabolites as a weak

CYP3A4 inducer was also supported by changes in plasma 4βHC, an emerging biomarker of

CYP3A4 activity (Bjorkhem-Bergman et al., 2013; Bodin et al., 2001; Leil et al., 2014). The net

effect was an increase in observed 4βHC, suggesting CYP3A4 induction. Based on the

relationships between midazolam AUC GMR, rifampicin dose, and plasma 4βHC, a framework Downloaded from

has been developed recently to classify the CYP3A4 inducer potencies of new chemical entities

after 14 days of dosing (Mangold et al., 2016). Using this framework (although not accepted dmd.aspetjournals.org currently by regulatory authorities) and considering the observed increase in plasma 4βHC in our

study (maximal 2-fold increase), midostaurin (100 mg bid for 28 days) could be viewed as a

moderate CYP3A4 inducer in the worst-case scenario. One caveat to using the plasma 4βHC at ASPET Journals on September 30, 2021

biomarker is that the potency classification can be dependent on sample size. Based on the low

variability of 4βHC, it is theorized that for a strong inducer such as rifampicin, there would be at

least an 80% probability of detecting a 4βHC elevation with a sample size of < 10. For detecting

moderate and weak inducers, the approximate sample size would be n = 10 to 25 and n > 60,

respectively (Leil et al., 2014). Given that the 4βHC data with midostaurin are based on only 10

patients, the classification of midostaurin as a weak to moderate CYP3A4 inducer is, at best,

supportive of the PBPK modeling results and needs further confirmation using a larger sample

size. Nonetheless, the 4βHC biomarker data provided an additional tool to evaluate the net effect

of CYP3A4 induction or inhibition by midostaurin and its metabolites at steady state when midostaurin was co-administered with midazolam and increased our confidence in the DDI predictions.

25

DMD Fast Forward. Published on November 8, 2017 as DOI: 10.1124/dmd.117.078006 This article has not been copyedited and formatted. The final version may differ from this version.

To evaluate the victim DDI potential of midostaurin, the CYP3A4 activity after single

and multiple doses of midostaurin needs to be considered. The change in simulated CYP3A4

activity with midostaurin alone and in combination with ketoconazole was similar in both single- and multiple-dose scenarios (Figure 4B). Based on this observation, CYP3A4 inhibitors are

predicted to have a similar impact on midostaurin exposure under single- and multiple-dose

conditions.

However, the DDI predictions in the presence of CYP3A4 inducers were found to be Downloaded from

highly dependent on single- vs multiple-dose conditions. As expected, CYP3A4 activity was not significantly affected by single-dose midostaurin, but it increased upon multiple dosing. dmd.aspetjournals.org Consequently, when another CYP3A4 inducer (rifampicin) was co-administered, the net increase

in CYP3A4 activity was less with a single dose than with multiple midostaurin doses, resulting

in a 90% decrease in midostaurin exposure under single-dose conditions vs a 43% decrease at ASPET Journals on September 30, 2021

under multiple-dose conditions. For these reasons, efavirenz (a moderate CYP3A4 inducer) was

predicted to have a minimal impact on midostaurin exposure at steady state (ie, a decrease of

8%).

It is noticed that the predicted increase in the AUC of midostaurin after co-administration of itraconazole 100 mg bid was lower than that with ketoconazole 400 mg qd. This is likely in part due to the metabolism of itraconazole as a CYP3A4 substrate being affected by midostaurin and its metabolites, resulting in the induction of CYP3A4 activity. The exposure of itraconazole in the presence of midostaurin was predicted to be lower than for itraconazole administered alone, and the inhibitory effect of itraconazole metabolites appears to be less potent than that of itraconazole alone.

26

DMD Fast Forward. Published on November 8, 2017 as DOI: 10.1124/dmd.117.078006 This article has not been copyedited and formatted. The final version may differ from this version.

Our simultaneous PBPK modeling approach takes the first step toward assessing the complex

CYP3A4 DDI potential of a parent drug and its 2 metabolites in healthy subjects. In order to assess the DDI potential of other CYP3A4 inhibitors/inducers used routinely in patients with cancer, two additional PBPK models were built for patients with AML and advSM. Patients with cancers are different than healthy subjects in terms of demographic and physiological properties.

Moreover, the population of patients with cancer is heterogeneous, and the differences in various characteristics for patients with cancer, as well as different types of cancer (eg, AML and Downloaded from advSM), can affect the ADME and PK of drugs. To fully characterize physiological differences between healthy subjects and patients with cancer, development of a cancer population file has dmd.aspetjournals.org been shown to be a promising tool for the prediction of PK in patients (Cheeti et al., 2013).

However, development, validation, and application of cancer-specific profiles will require more comprehensive data collection. Due to the limited knowledge and understanding of the at ASPET Journals on September 30, 2021 mechanisms of the PK differences observed in healthy subjects and patients with cancer, two individual compound files were built for patients with AML and patients with advSM; the lower

Vss and CLint of midostaurin for patients with both cancer and lower clearance of CGP52421 for

AML compared with the model for healthy subjects were applied to the patient models. Lower

clearances of midostaurin and CGP52421 in patients with AML than in healthy subjects are

likely due to multiple factors. The mechanism of the differences in these parameters is not clear;

however, it may be a difference in midostaurin distribution (although similar plasma protein

binding between healthy subjects and patients was observed in ex vivo studies) and metabolic

clearance between the two populations. It could also be that the metabolic clearance of

CGP52421 in patients with AML is different than that in healthy subjects and patients with

advSM. In recent years, researchers have suggested that circulating levels of cytokines, including

27

DMD Fast Forward. Published on November 8, 2017 as DOI: 10.1124/dmd.117.078006 This article has not been copyedited and formatted. The final version may differ from this version.

IL-6, are significantly higher in patients with AML than in healthy subjects (Tsimberidou et al.,

2008; Meyers et al., 2005). Furthermore, suppression of CYP3A4 by IL-6 in hepatocytes was

observed and clinical examples have been reported showing that CYP3A4-mediated metabolism

was affected by cytokine modulators (eg, elevation of IL-6 level), resulting in reduced CYP3A4

expression (Evers et al., 2013). In addition, a lower Fa of midostaurin than those in the healthy

subject model was used for the advSM model. The reduced overall absorption in patients with advSM could be due to the disease-related abnormal numbers of mast cells in organs, including Downloaded from the gastrointestinal tract (Cardet et al., 2013), which may affect oral absorption.

In conclusion, the PBPK model described here using a middle-out approach reasonably dmd.aspetjournals.org predicted PK profiles of midostaurin and its 2 metabolites after single and multiple dosing. The simultaneous parent and metabolite modeling approach allowed predictions under steady-state conditions that were not possible to achieve in healthy subjects. The model also captured the at ASPET Journals on September 30, 2021 apparent effect of mixed TDI/induction mechanisms. Our combined, multipronged approach allowed the extension of findings from single-dose clinical DDI studies and enabled predictions of steady-state DDI effects to be made with improved confidence. The results generated using this integrated approach provide support of clinical recommendations and potential product label language.

ACKNOWLEDGMENTS

The data analysis of 4βHC was conducted by statistician Matthieu Villeneuve, a former

employee of Novartis. Medical editorial assistance was provided by Jennifer Gooch, PhD, and

Pamela Tuttle, PhD, CMPP, of ArticulateScience LLC. Funding for medical editorial assistance

was provided by Novartis Pharmaceuticals Corporation.

28

DMD Fast Forward. Published on November 8, 2017 as DOI: 10.1124/dmd.117.078006 This article has not been copyedited and formatted. The final version may differ from this version.

AUTHORSHIP CONTRIBUTIONS

Participated in research design: Gu, Dutreix, Einolf, He

Conducted experiments: Gu, Chun, Wang

Performed data analysis: Gu, Dutreix, Villeneuve

Contributed to the writing of the manuscript: Gu, Rebello, Dutreix, Einolf, He, Ouatas Downloaded from

dmd.aspetjournals.org at ASPET Journals on September 30, 2021

29

DMD Fast Forward. Published on November 8, 2017 as DOI: 10.1124/dmd.117.078006 This article has not been copyedited and formatted. The final version may differ from this version.

REFERENCES

Andrejauskas-Buchdunger E and Regenass U (1992) Differential inhibition of the epidermal

growth factor-, platelet-derived growth factor-, and protein kinase C-mediated signal

transduction pathways by the staurosporine derivative CGP 41251. Cancer Res 52: 5353–

5358.

Barry EV, Clark JJ, Cools JV, Roessel J, and Gilliland DG (2007) Uniform sensitivity of FLT3 Downloaded from activation loop mutants to the tyrosine kinase inhibitor midostaurin. Blood 110: 4476–4479.

Bjorkhem-Bergman L, Bäckström T, Nylén H, Rönquist-Nii Y, Bredberg E, Andersson TB,

Bertilsson L, and Diczfalusy U (2013) Comparison of endogenous 4beta-hydroxycholesterol dmd.aspetjournals.org

with midazolam as markers for CYP3A4 induction by rifampicin. Drug Metab Dispos 41:

1488–1493.

Bodin K, Bretillon L, Aden Y, Bertilsson L, Broomé U, Einarsson C, and Diczfalusy U (2001) at ASPET Journals on September 30, 2021

Antiepileptic drugs increase plasma levels of 4beta-hydroxycholesterol in humans: evidence

for involvement of cytochrome p450 3A4. J Biol Chem 276: 38685–38689.

Cardet JC, Akin C, and Lee MJ (2013) Mastocytosis: update on pharmacotherapy and future

directions. Expert Opin Pharmacother 14: 2033–2045.

Cheeti S, Budha NR, Rajan S, Dresser MJ, and Jin JY (2013) A physiologically based

pharmacokinetic (PBPK) approach to evaluate pharmacokinetics in patients with

cancer. Biopharm Drug Dispos 34: 141–154.

Committee for Human Medicinal Products (2012) Guideline on the Investigation of Drug

Interactions. European Medicines Agency, London.

Diczfalusy U, Nylen H, Elander P, and Bertilsson L (2011) 4beta-hydroxycholesterol, an

endogenous marker of CYP3A4/5 activity in humans. Br J Clin Pharmacol 71: 183–189.

30

DMD Fast Forward. Published on November 8, 2017 as DOI: 10.1124/dmd.117.078006 This article has not been copyedited and formatted. The final version may differ from this version.

Dutreix C, Munarini F, Lorenzo S, Roesel J, and Wang Y (2013) Investigation into CYP3A4-

mediated drug-drug interactions on midostaurin in healthy volunteers. Cancer Chemother

Pharmacol 72: 1223–1234.

Dutreix C, Sebastien L, and Wang Y (2014) Comparison of two endogenous biomarkers of

CYP3A4 activity in a drug-drug interaction study between midostaurin and rifampicin. Eur J

Clin Pharmacol 70: 915–920.

Elias A, Wu J, Chen T (2013). Tumor suppressor protein p53 negatively regulates human Downloaded from

pregnane X receptor activity. Mol Pharmacol 83: 1229–1236.

European Medicine Agency (EMA), Committee for Human Medicinal Products (CHMP) dmd.aspetjournals.org Guideline on the investigation of drug interactions. 21 June 2012.

Fabbro D, Ruetz S, Bodis S, Pruschy M, Csermak K, Man A, Campochiaro P, Wood J, O’Reilly

T, and Meyer T (2000) PKC412: a protein kinase inhibitor with a broad therapeutic potential. at ASPET Journals on September 30, 2021

Anticancer Drug Des 15: 17–28.

Fahmi OA, Hurst S, Plowchalk D, Cook J, Guo F, Youdim K, Dickins M, Phipps A, Darekar A,

Hyland R, and Obach RS (2009) Comparison of different algorithms for predicting clinical

drug–drug interactions, based on the use of CYP3A4 in vitro data: predictions of compounds

as precipitants of interaction. Drug Metab Dispos 37: 1658–1666.

FDA Guidance for Industry (2012) Drug Interaction Studies—Study Design, Data Analysis,

Implications for Dosing, and Labeling Recommendations. US Department of Health and

Human Services, Food and Drug Administration, Center for Drug Evaluation and Research,

Center for Veterinary Medicine, Rockville, MD.

31

DMD Fast Forward. Published on November 8, 2017 as DOI: 10.1124/dmd.117.078006 This article has not been copyedited and formatted. The final version may differ from this version.

Fenneteau F, Poulin P, and Nekka F (2010) Physiologically based predictions of the impact of

inhibition of intestinal and hepatic metabolism on human pharmacokinetics of CYP3A

substrates. J Pharm Sci 99: 486–514.

Fischer T, Stone RM, Deangelo DJ, Galinsky I, Estey E, Lanza C, Fox E, Ehninger G, Feldman

EJ, Schiller GJ, Klimek VM, Nimer SD, Gilliland DG, Dutreix C, Huntsman-Labed A,

Virkus J, and Giles FJ (2010) Phase IIB trial of oral midostaurin (PKC412), the FMS-like

tyrosine kinase 3 receptor (FLT3) and multi-targeted kinase inhibitor, in patients with acute Downloaded from

myeloid leukemia and high-risk myelodysplastic syndrome with either wild-type or mutated

FLT3. J Clin Oncol 28: 4339–4345. dmd.aspetjournals.org Galetin A, Burt H, Gibbons L, and Houston JB (2006) Prediction of time-dependent CYP3A4

drug–drug interactions: impact of enzyme degradation, parallel elimination pathways, and

intestinal inhibition. Drug Metab Dispos 34: 166–175. at ASPET Journals on September 30, 2021

Gallogly MM and Lazarus HM (2016) Midostaurin: an emerging treatment for acute myeloid

leukemia patients. J Blood Metabol 7: 73–83.

Garg V, Chandorkar G, Farmer HF, Smith F, Alves K, and van Heeswijk RP (2012) Effect of

telaprevir on the pharmacokinetics of midazolam and digoxin. J Clin Pharmacol 52: 1566–

1573.

Goldstein I, Rivlin N, Shoshana OY, Ezra O, Madar S, Goldfinger N, Rotter V (2013).

Chemotherapeutic agents induce the expression and activity of their clearing enzyme

CYP3A4 by activating p53. Carcinogenesis 34: 190–198.

Gotlib J, Berube C, Growney JD, Smith F, Alves K, and van Heeswijk RP (2005) Activity of the

tyrosine kinase inhibitor PKC412 in a patient with mast cell leukemia with the D816V KIT

mutation. Blood 106: 2865–2870.

32

DMD Fast Forward. Published on November 8, 2017 as DOI: 10.1124/dmd.117.078006 This article has not been copyedited and formatted. The final version may differ from this version.

Gotlib J, Kluin-Nelemans H, George TI, Akin C, Sotlar K, Hermine O, Awan FT, Hexner E,

Mauro MJ, Sternberg DW, Villeneuve M, Huntsman Labed A, Stanek EJ, Hartmann K,

Horny HP, Valent P, and Reiter A (2016) Efficacy and safety of midostaurin in advanced

systemic mastocytosis. N Engl J Med 374: 2530–2541.

Harmsen S, Meijerman I, Beijnen JH, Schellens JH (2008). Nuclear receptor mediated induction

of cytochrome P450 3A4 by anticancer drugs: a key role for the pregnane X receptor. Cancer

Chemother Pharmacol 64: 35–43. Downloaded from

He H, Tran P, Gu H, Tedesco V, Zhang J, Lin W, Gatlik E, Klein K, and Heimbach T (2017)

Midostaurin, a novel protein kinase inhibitor for the treatment of acute myelogenous dmd.aspetjournals.org leukemia: insights from human absorption, metabolism, and excretion studies of a BDDCS II

drug. Drug Metab Dispos 45: 540–555.

Leil TA, Kasichayanula S, Boulton DW, and LaCreta F (2014) Evaluation of 4beta- at ASPET Journals on September 30, 2021

hydroxycholesterol as a clinical biomarker of CYP3A4 drug interactions using a Bayesian

mechanism-based pharmacometric model. CPT Pharmacometrics Syst Pharmacol. 3: e120.

Levis M, Brown P, Smith D, Stine A, Pham R, Stone R, Deangelo D, Galinsky I, Giles F, Estey

E, Kantarjian H, Cohen P, Wang Y, Roesel J, Karp JE, and Small D (2006) Plasma inhibitory

activity (PIA): a pharmacodynamic assay reveals insights into the basis for cytotoxic

response to FLT3 inhibitors. Blood 18: 3477–3483.

Lin Z, Fahmi OA, Johnson N, Rodrigues DA, Obach RS, Steyn SJ, Zientek M, and Goosen

TC. (2016) Tyrosine kinase inhibitors robustly induced CYP3A4 mRNA, but not

activity via multiple pathways. Poster presented at: 21st International Symposium on

Microsomes and Drug Oxidations; October 2–6, 2016; Davis, CA.

33

DMD Fast Forward. Published on November 8, 2017 as DOI: 10.1124/dmd.117.078006 This article has not been copyedited and formatted. The final version may differ from this version.

Mangold JB, Wu F, and Rebello S (2016) Compelling relationship of CYP3A induction to levels

of the putative biomarker 4β-hydroxycholesterol and changes in midazolam exposure. Clin

Pharm Drug Develop 5: 245–249.

Meyers CA, Albitar M, Estey E (2005). Cognitive impairment, fatigue, and cytokine levels in

patients with acute myelogenous leukemia or myelodysplastic syndrome. Cancer 104: 788–

793.

Propper DJ, McDonald AC, Man A, Thavasu P, Balkwill F, Braybrooke JP, Caponigro F, Graf Downloaded from

P, Dutreix C, Blackie R, Kaye SB, Ganesan TS, Talbot DC, Harris AL, and Twelves C

(2001) Phase I and pharmacokinetic study of PKC412, an inhibitor of protein kinase C. J dmd.aspetjournals.org Clin Oncol 19: 1485–1492.

Prueksaritanont T, Chu X, Gibson C, Cui D, Yee KL, Ballard J, Cabalu T, and Hochman J

(2013) Drug–drug interaction studies: regulatory guidance and an industry perspective. AAPS at ASPET Journals on September 30, 2021

J 15: 629–645.

Reitman ML, Chu X, Cai X, Yabut J, Venkatasubramanian R, Zajic S, Stone JA, Ding Y, Witter

R, Gibson C, Roupe K, Evers R, Wagner JA, and Stoch A (2011) Rifampin’s acute inhibitory

and chronic inductive drug interactions: experimental and model-based approaches to drug–

drug interaction trial design. Clin Pharmacol Ther 89: 234–242.

Stone RM, DeAngelo DJ, Klimek V, Galinsky I, Estey E, Nimer SD, Grandin W, Lebwohl D,

Wang Y, Cohen P, Fox EA, Neuberg D, Clark J, Gilliland DG, and Griffin JD (2005)

Patients with acute myeloid leukemia and an activating mutation in FLT3 respond to a small-

molecule FLT3 tyrosine kinase inhibitor, PKC412. Blood 105: 54–60.

Stone RM, Mandrekar S, Sandord, L, Geyer S, Bloomfield CD, Döhner K, Thiede C, Marcucci

G, Lo-Coco F, Klisovic RB, Wei A, Sierra J, Sanz MA, Brandwein JM, de Witte T,

34

DMD Fast Forward. Published on November 8, 2017 as DOI: 10.1124/dmd.117.078006 This article has not been copyedited and formatted. The final version may differ from this version.

Niederwieser D, Appelbaum FR, Medeiros BC, Tallman MS, Krauter J, Schlenk RF, Ganser

A, Serve H, Ehninger G, Amadori S, Larson RA, and Döhner H (2015) The multi-kinase

inhibitor midostaurin (M) prolongs survival compared with placebo (P) in combination with

daunorubicin (D)/cytarabine (C) induction (ind), high-dose C consolidation (consol), and as

maintenance (maint) therapy in newly diagnosed acute myeloid leukemia (AML) patients

(pts) age 18-60 with FLT3 mutations (muts): an international prospective randomized (rand)

P-controlled double-blind trial (CALGB 10603/RATIFY [Alliance]). Blood 126: abstract 6. Downloaded from

Tsimberidou AM, Estey E, Wen S, Pierce S, Kantarjian H, Albitar M, Kurzrock R (2008). The

prognostic significance of cytokine levels in newly diagnosed acute myeloid leukemia and dmd.aspetjournals.org high-risk myelodysplastic syndromes. Cancer 113: 1605–1613.

Wang Y, Yin OQ, Graf P, Kisicki JC, and Schran H (2008) Dose- and time-dependent

pharmacokinetics of midostaurin in patients with diabetes mellitus. J Clin Pharmacol 48: at ASPET Journals on September 30, 2021

763–775.

Weisberg E, Boulton C, Kelly LM, Manley P, Fabbro D, Meyer T, Gilliland DG, and Griffin JD

(2002) Inhibition of mutant FLT3 receptors in leukemia cells by the small molecule tyrosine

kinase inhibitor PKC412. Cancer Cell 1: 433–443.

Weiss HM, Gatlik E (2014). Equilibrium gel filtration to measure plasma protein binding of very

highly bound drugs. J Pharm Sci. 103:752-759.

Yeo KR, Walsky RL, Jamei M, Rostami-Hodjegan A, and Tucker GT (2011) Prediction of time-

dependent CYP3A4 drug–drug interactions by physiologically based pharmacokinetic

modelling: impact of inactivation parameters and enzyme turnover. Eur J Pharm Sci 43:

160–173.

35

DMD Fast Forward. Published on November 8, 2017 as DOI: 10.1124/dmd.117.078006 This article has not been copyedited and formatted. The final version may differ from this version.

Yin OQ, Wang Y, and Schran H (2008) A mechanism-based population pharmacokinetic model

for characterizing time-dependent pharmacokinetics of midostaurin and its metabolites in

human subjects. Clin Pharmacokinet 47: 807–816.

Downloaded from dmd.aspetjournals.org at ASPET Journals on September 30, 2021

36

DMD Fast Forward. Published on November 8, 2017 as DOI: 10.1124/dmd.117.078006 This article has not been copyedited and formatted. The final version may differ from this version.

FOOTNOTES

Funding for this project was provided by Novartis.

Downloaded from dmd.aspetjournals.org at ASPET Journals on September 30, 2021

37

DMD Fast Forward. Published on November 8, 2017 as DOI: 10.1124/dmd.117.078006 This article has not been copyedited and formatted. The final version may differ from this version.

FIGURE LEGENDS

Figure 1. The black lines represent the simulated mean plasma concentration-time profiles of

midostaurin (panels A, D, and G), CGP52421 (panels B, E, and H), and CGP62221 (panels C, F,

and I). The dashed lines represent the simulated lower 5th and upper 95th percentiles. The

symbols and error bars represent the observed mean plasma concentration-time data and standard

deviation from 2 clinical trials (A2111 and A2108) and 2 control trials (A2109 and A2110) for Downloaded from panels A, B, and C. All 4 trials were open-label, randomized, parallel-group studies in healthy volunteers. Panels A, B, and C show day 1 following midostaurin 50 mg; panels D, E, and F

show days 1 to 3 following midostaurin 75 mg twice daily for 3 days; and panels G, H, and I dmd.aspetjournals.org show days 1 and 7 following midostaurin 50 mg twice daily for 6 days and a single dose on day

7.

Figure 2. The simulated mean plasma concentration-time profiles of midostaurin after treatment at ASPET Journals on September 30, 2021 with midostaurin 50 mg (A) on day 6 in the absence (solid line) or presence (dashed line) of ketoconazole 400 mg once daily on days 1 to 10 and (B) on day 9 in the absence (solid line) or presence (dashed line) of rifampicin 600 mg once daily on days 1 to 14. The symbols (with and without interaction in red and purple, respectively) and error bars represent the observed mean plasma concentration-time data and standard deviation, respectively.

Figure 3. Simulated area under the curve (AUC) and maximum concentration (Cmax) ratios (with

90% CIs) in healthy volunteers (HVs) and in patients with acute myeloid leukemia (AML) and

advanced systemic mastocytosis (advSM) for (A) midostaurin, (B) CGP52421, and (C)

CGP62221 when co-administered with ketoconazole, itraconazole, fluconazole, efavirenz, and

rifampicin. The open circles and closed squares represent geometric mean Cmax and AUC ratios,

respectively.

38

DMD Fast Forward. Published on November 8, 2017 as DOI: 10.1124/dmd.117.078006 This article has not been copyedited and formatted. The final version may differ from this version.

Figure 4. (A) The solid green and red lines represent the simulated cytochrome P450 3A4

(CYP3A4) hepatic activity (% of baseline) after a 50-mg single dose and a 50-mg twice-daily dose of midostaurin to steady state (day 28), respectively, after treatment with rifampicin 600 mg once daily. (B) The solid green and red lines represent the simulated CYP3A4 hepatic activity

(% of baseline) after a 50-mg single dose and a 50-mg twice-daily dose of midostaurin to steady state (day 28), respectively, after treatment with ketoconazole 400 mg once daily.

Figure 5. The solid lines represent the simulated mean systemic plasma concentration-time Downloaded from profiles of midazolam 4 mg (A) in the absence of midostaurin, (B) with a single dose of midostaurin 100 mg on day 1, and (C) with an additional twice-daily dose of midostaurin 50 mg dmd.aspetjournals.org on days 2 to 4. The dashed lines represent the simulated lower 5th and upper 95th percentiles.

The symbols (with and without interaction in red and purple, respectively) and error bars represent the observed mean plasma concentration-time data and standard deviation, at ASPET Journals on September 30, 2021 respectively.

Figure 6. The lines represent the mean plasma concentration-time profiles of (A) midazolam,

(B) midostaurin, (C) CGP52421, and (D) CGP62221 following co-administration of midazolam

4 mg and midostaurin 50 mg twice daily for 28 days.

Figure 7. Overlays of measured 4β-hydroxycholesterol (4βHC) profiles with ratios relative to baseline and predicted changes in midazolam area under the curve (AUC) ratios affected by midostaurin and its metabolites. Negative control: healthy volunteers who received placebo on days 1 to 15 and midostaurin on day 9. Positive control: healthy volunteers who received rifampicin 600 mg once daily on days 1 to 15 and midostaurin on day 9. Midostaurin: patients with advanced systemic mastocytosis who received midostaurin 100 mg twice daily on days 1 to

28.

39

DMD Fast Forward. Published on November 8, 2017 as DOI: 10.1124/dmd.117.078006 This article has not been copyedited and formatted. The final version may differ from this version.

GM, geometric mean.

Downloaded from dmd.aspetjournals.org at ASPET Journals on September 30, 2021

40

DMD Fast Forward. Published on November 8, 2017 as DOI: 10.1124/dmd.117.078006 This article has not been copyedited and formatted. The final version may differ from this version.

Downloaded from Table 1. Input Parameters for Midostaurin, CGP52421, and CGP62221

Parameter Midostaurin CGP52421 CGP62221

Value Reference Value Reference Value dmd.aspetjournals.org Reference

MW (g/mol) 570.6 586.6 556.6 logPo:w 5.49 Internal database 4.76 Internal database 4.68 Internal database pKa 11.2 Predicteda 10.8 Predicteda 11.2 Predicteda at ASPET Journals on September 30, 2021

Compound type Monoprotic base fup 0.00015 Internally measured 0.00021 Internally measured 0.00038 Internally measured

B/P 0.55 0.55 Assumed same as parent 0.55 Assumed same as parent

Plasma-binding AGP AGP AGP component

First-order absorption

Fa 0.85 User defined (optimized)

(0.65, advSM)

−1 ka (h ) 1.5

(0.683, advSM)

Lag time (h) 0.3

Qgut (L/h) 5.23 Simcyp predicted fugut 0.3

41

DMD Fast Forward. Published on November 8, 2017 as DOI: 10.1124/dmd.117.078006 This article has not been copyedited and formatted. The final version may differ from this version.

Downloaded from Parameter Midostaurin CGP52421 CGP62221

Value Reference Value Reference Value Reference

Peff, man (10−4 cm/s) 0.835 dmd.aspetjournals.org

Caco-2 (10−6 cm/s) 1.4 Internally measured

Caco-2 reference (10−6 17 cm/s) at ASPET Journals on September 30, 2021 Minimal with single adjusting compartmental distribution

Q (L/h) 3 Estimated from clinical PK 10 Estimated from clinical 2 Estimated from clinical PK

(2.889, AML/ data (a single-dose trial) PK data (a single-dose data (a single-dose trial)

advSM) trial)

Volume (Vsac [L/h]) 0.82 1.8 1.1

(0.623,

AML/advSM)

Vss (L/h) 1 1.3 1.3

(0.742,

AML/advSM)

Elimination

Hepatic CL by CYP3A4 100 Estimated from human ADME 90 Estimated from human 90 Estimated from human

(%) study ADME study ADME study

CLint, CYP3A4 9.3 See Elimination

42

DMD Fast Forward. Published on November 8, 2017 as DOI: 10.1124/dmd.117.078006 This article has not been copyedited and formatted. The final version may differ from this version.

Downloaded from Parameter Midostaurin CGP52421 CGP62221

Value Reference Value Reference Value Reference

formation of CGP52421 (3.9, dmd.aspetjournals.org

(µL/min/pmol CYP3A4) AML/advSM)

CLint, CYP3A4 9.3 formation of CGP62221 (3.9, at ASPET Journals on September 30, 2021 (µL/min/pmol CYP3A4) AML/advSM)

CLint, CYP3A4 6.6 3.18 1.19 formation of other (2.8, (0.557, metabolites AML/advSM) AML

(µL/min/pmol CYP3A4) patients)

Additional HLM 48.4 18.1 clearance (µL/min/mg (6.48, protein) AML

patients)

Active uptake into 1 Default hepatocytes

CLR 0 See Elimination

CYP3A4-related interaction

43

DMD Fast Forward. Published on November 8, 2017 as DOI: 10.1124/dmd.117.078006 This article has not been copyedited and formatted. The final version may differ from this version.

Downloaded from Parameter Midostaurin CGP52421 CGP62221

Value Reference Value Reference Value Reference

Reversible inhibition 0.25 Internally measured 0.44 Internally measured 0.25 dmd.aspetjournals.org Internally measured

CYP3A4 Kiu (µM) (Supplemental Table 2) (Supplemental Table 2) (Supplemental Table 2)

TDI

Internally measured Internally measured at ASPET Journals on September 30, 2021 Internally measured KI, total (μM) 1.02 1.97 2.01 (Supplemental Table 3) (Supplemental Table 3) (Supplemental Table 3) kinact (1/h) 2.8 3.0 4.62

Induction of CYP3A4 9.14 Internally measured 10.8 Internally measured 11.97 Internally measured

Indmax (fold) 0.0026 (Supplemental Table 4) and 0.0053 (Supplemental Table 4) 0.0027 (Supplemental Table 4)

IndC50 (µM) optimized and optimized and optimized

a Determined using the ADMET Predictor 6.5, Simulations Plus, Lancaster, CA).

ADME, absorption, distribution, metabolism, and excretion; advSM, advanced systemic mastocytosis; AGP, α1-acid glycoprotein; AML, acute myeloid leukemia; B/P, blood-to-plasma ratio; Caco-2, continuous heterogeneous human epithelial colorectal adenocarcinoma cell line; CL, clearance; CLint, intrinsic clearance; CLR, renal clearance; CYP3A4, cytochrome P450 3A4; IndC50, concentration of inducer at half maximal induction; Fa, fraction of dose absorbed; fugut, fraction of unbound drug in the gut; fup, fraction of unbound drug in plasma; HLM, human liver microsome; Ind50, half-

maximum induction rate; Indmax, maximal fold induction over vehicle control; ka, absorption rate constant; KI, concentration producing a half-maximal rate of activity reduction; kinact, maximal rate of

activity reduction; Kiu, unbound inhibition constant; logPo:w, water partition coefficient; MW, molecular weight; Peff, man, effective permeability in man; pKa, acid dissociation constant; PK,

pharmacokinetics; Q, intercompartmental clearance; Qgut, nominal flow through the gut; Vsac, volume of distribution for the single adjustable compartment; Vss, volume of distribution at steady state.

44

DMD Fast Forward. Published on November 8, 2017 as DOI: 10.1124/dmd.117.078006 This article has not been copyedited and formatted. The final version may differ from this version.

Table 2. Summary of Trial Simulation Design

Simulated Observed PK Measurement

Population Population Dosing Regimen of Substrate

Model performance of Healthy Healthy Ketoconazole 400 mg qd on 0-120 h, 0-inf midostaurin PK and volunteer volunteer days 1-10 + midostaurin ketoconazole DDIs (FMI) 50 mg on day 6

Model performance of Healthy Healthy Rifampicin 600 mg qd on 0-144 h, 0-inf Downloaded from midostaurin PK and rifampicin volunteer volunteer days 1-14 + midostaurin

DDIs (FMI) 50 mg on day 9

Model performance of Healthy Healthy Midazolam 4 mg qd on Day 1: 0-10 h, 0-inf dmd.aspetjournals.org midazolam PK and midostaurin- volunteer volunteer days 1 and 6 + midostaurin Day 6: 0-10 h midazolam DDIs 100-mg single dose on day

1, 50 mg bid on days 2-4 at ASPET Journals on September 30, 2021 bid, twice daily; DDI, drug-drug interaction; FMI, final market image; inf, infinity; PK, pharmacokinetics; qd, once daily.

45

DMD Fast Forward. Published on November 8, 2017 as DOI: 10.1124/dmd.117.078006 This article has not been copyedited and formatted. The final version may differ from this version.

Table 3. 4βHC Study Design (CPKC412A2110; registered as EudraCT no. 2009-009895-11)

Population Study Arm Treatment Screening Study Day

1

) − 1 (

to

14 27 21 7 - - - 14 2 3 8 9 10 11 15 28 22 4 −

12 16 - 23 s ay Baseline D

Healthy Negative Placebo qd volunteers control Midostaurin 50 mg

(n = 20)

bid Downloaded from

Rifampicin 600 mg Positive qd control Midostaurin 50 mg

(n = 20) dmd.aspetjournals.org bid

PK 4βHC a a a a assessment

Patients with Midostaurin Midostaurin 100 mg at ASPET Journals on September 30, 2021

advSM arm (n = 10) bid

PK 4βHC b b b b b b assessment

Light-gray highlight indicates receipt of placebo. Medium-gray highlight indicates treatment with rifampicin (a CYP3A4 inducer). Black highlight indicates treatment with midostaurin.

4βHC, 4β-hydroxycholesterol; advSM, advanced systemic mastocytosis; bid, twice daily; PK, pharmacokinetics; qd, once daily.

Blood samples for the assessment of 4βHC in serum were taken as follows: a Healthy volunteers: days 1, 9 (before midostaurin treatment), 11, and 15. b Patients with advSM: days 1, 3, 8, 15, 22, and 28.

46

DMD Fast Forward. Published on November 8, 2017 as DOI: 10.1124/dmd.117.078006 This article has not been copyedited and formatted. The final version may differ from this version.

Table 4. Summary of Predicted and Observed AUC and Cmax Ratios With Ketoconazole and

Rifampicin

Ketoconazole 400 mg AUC0-120h GMR Cmax GMR With Co- qd on Days 1-10 + (90% CI range) (90% CI range) Administration Midostaurin 50 mg of Ketoconazole Observed Predicted Observed Predicted on Day 6

6.1 5.8 1.8 2.1 Midostaurin Downloaded from (5.0-7.5) (5.1-6.5) (1.6-2.1) (1.9-2.2)

1.2 0.6 0.5 0.4 CGP52421

(1.0-1.5) (0.5-0.7) (0.4-0.6) (0.4-0.4) dmd.aspetjournals.org

1.0 0.6 0.6 0.3 CGP62221 (0.8-1.2) (0.6-0.7) (0.5-0.7) (0.3-0.3)

Rifampicin 600 mg AUC0-144h GMR Cmax GMR at ASPET Journals on September 30, 2021 With Co- qd on Days 1-14 + (90% CI range) (90% CI range) Administration Midostaurin 50 mg of Rifampicin Observed Predicted Observed Predicted on Day 9

0.06 0.10 0.27 0.21 Midostaurin (0.05-0.07) (0.09-0.12) (0.23-0.31) (0.18-0.25)

0.41 0.35 0.65 0.91 CGP52421 (0.36-0.46) (0.31-0.40) (0.59-0.72) (0.84-0.99)

0.08 0.45 0.63 1.02 CGP62221 (0.06-0.09) (0.40-0.50) (0.56-0.70) (0.97-1.09)

AUC, area under the curve; Cmax, maximum concentration; GMR, geometric mean ratio; qd, once daily.

47

DMD Fast Forward. Published on November 8, 2017 as DOI: 10.1124/dmd.117.078006 This article has not been copyedited and formatted. The final version may differ from this version.

Table 5. Geometric Mean 4βHC Plasma Levels at Each Time Point Midostaurin

Negative Control Positive Control (n = 10; 4 males and

(n = 20) (n = 20) 6 females)

GM 4βHC Plasma Levels (CV %) (ng/mL)

Baseline (day 1) 25.3 (35.5) 22.0 (44.0) 29.2 (43.7)

Day 3 NA NA 34.1 (43.7) Downloaded from Day 8 NA NA 41.7 (39.1)

Day 9 23.4 (36.4) 75.0 (32.1) NA

Day 11 25.4 (35.9) 89.5 (33.5) NA dmd.aspetjournals.org

Day 15 23.3 (38.8) 102.7 (27.6) 53.8 (34.7)

Day 22 NA NA 51.4 (37.1)

Day 28 NA NA 56.0 (35.6) at ASPET Journals on September 30, 2021

GM Percent Change in 4βHC Plasma Levels From Baseline (CV %) (%)

Day 3 NA NA 16.8 (0.1)

Day 8 NA NA 42.9 (−10.3)

Day 9 −7.7 (102) 240.3 (−26.9) NA

Day 11 0.4 (101) 306.0 (−23.9) NA

Day 15 −8.1 (109) 366.1 (−37.3) 84.2 (−20.6)

Day 22 NA NA 76.1 (−14.9)

Day 28 NA NA 91.7 (18.4)

4βHC, 4β-hydroxycholesterol; CV, coefficient of variation; GM, geometric mean; NA, not applicable.

48

DMD Fast Forward. Published on November 8, 2017 as DOI: 10.1124/dmd.117.078006 This article has not been copyedited and formatted. The final version may differ from this version. Downloaded from dmd.aspetjournals.org

A B C 5000 Midostaurin 1600 CGP52421 2500 CGP62221 at ASPET Journals on September 30, 2021 Midostaurin 95th percentile CGP52421 95th percentile CGP62221 95th percentile 4500 Midostaurin 5th percentile 1400 CGP52421 5th percentile CGP62221 5th percentile

(ng /mL) 4000 A2111 A2111 2000 A2111 1200 on 3500 A2108 A2108 A2108 A2109 control arm A2109 control arm A2109 control arm 1000 3000 A2110 control arm A2110 control arm 1500 A2110 control arm 2500 800 once ntrati 2000 1000 C 600 1500 400 1000 500 200

do staurin 500 CGP62221 Concentration (ng/mL) CGP52421 Concentration (ng/mL) Mi 0 0 0 0 6 12 18 24 30 36 42 48 0 6 12 18 24 30 36 42 48 0 6 12 18 24 30 36 42 48 Time (hours) Time (hours) Time (hours)

Midostaurin CGP52421 CGP62221 D Midostaurin 95th percentile E CGP52421 95th percentile F CGP62221 95th percentile Midostaurin 5th percentile CGP52421 5th percentile CGP62221 5th percentile 9000 A2113 5000 A2113 6000 A2113 8000 4500 5000 (ng /mL) 7000 4000

on 6000 3500 4000 5000 3000 2500 3000 4000 once ntrati

C 2000 3000 2000 1500 2000 1000 1000 1000 500 do staurin CGP52421 Concentration (ng/mL) CGP62221 Concentration (ng/mL) Mi 0 0 0 0 8 16 24 32 40 48 56 64 72 0 8 16 24 32 40 48 56 64 72 0 8 16 24 32 40 48 56 64 72 Time (hours) Time (hours) Time (hours)

Midostaurin CGP52421 CGP62221 G Midostaurin 95th percentile H CGP52421 95th percentile I CGP62221 95th percentile 6000 Midostaurin 5th percentile 3500 CGP52421 5th percentile 4500 CGP62221 5th percentile A2116 healthy volunteers A2116 healthy volunteers A2116 healthy volunteers 4000 5000 3000 (ng /mL) 3500

on 2500 4000 3000 2000 2500 3000 2000 once ntrati 1500 C 2000 1500 1000 1000 1000 500 500 do staurin CGP62221 CGP62221 Concentration (ng/mL) CGP52421 CGP52421 Concentration (ng/mL) Mi 0 0 0 0 4 8 12 16 20 24 144 148 152 156 160 164 168 0 4 8 12 16 20 24 144 148 152 156 160 164 168 0 4 8 12 16 20 24 144 148 152 156 160 164 168 Time (hours) Time (hours) Time (hours)

Figure 1 DMD Fast Forward. Published on November 8, 2017 as DOI: 10.1124/dmd.117.078006 This article has not been copyedited and formatted. The final version may differ from this version. Downloaded from dmd.aspetjournals.org at ASPET Journals on September 30, 2021

A 4000 B 2500 Midostaurin Midostaurin Midostaurin with ketoconazole Midostaurin with rifampicin 3500 Observed (without ketoconazole) Observed (without rifampicin) 2000

(ng /mL) Observed (with ketoconazole) Observed (with rifampicin) 3000 (ng /mL)

2500 1500

2000 1000 1500

1000 500 500 staurin Mi do staurin on C on centrati staurin Mi do staurin on C on centrati

0 0 96 108 120 132 144 156 174 186 198 210 222 Time (hours) Time (hours) Figure 2 DMD Fast Forward. Published on November 8, 2017 as DOI: 10.1124/dmd.117.078006 This article has not been copyedited and formatted. The final version may differ from this version. Downloaded from

A dmd.aspetjournals.org Predicted C max Predicted C max Cmax 3.0 Predicted AUC Predicted AUC Unity AUC 5.4 Ketoconazole HVs AML/advSM Cmax 2.6/3.1 AUC 4.2/4.4

Cmax 1.7 AUC 2.7 Fluconazole Cmax 1.7 AUC 2.5/2.6 on s ond iti Cmax 1.7 AUC 2.7 Itraconazole Cmax 1.6/1.7 AUC 2.2/2.0

Cmax 0.95

tment C ea tment AUC 0.92

Efavirenz at ASPET Journals on September 30, 2021 Tr Cmax 0.97/0.98 AUC 0.95/0.97

Cmax 0.70 AUC 0.57 Rifampicin Cmax 0.78/0.76 AUC 0.63/0.66

Inducer Inhibitor

Strong Moderate Weak Weak Moderate Strong

0.01 0.1 1 10 100 Effect Size (log fold change) B Predicted C Predicted C max max Cmax 1.5

Predicted AUC Predicted AUC Unity AUC 1.6 HVs AML/advSM Cmax 1.1/1.5 Ketoconazole AUC 1.1/1

Cmax 1.4 AUC 1.6 Fluconazole Cmax 1.1/1.5 AUC 1.2/1.6 on s ond iti Cmax 1.2 AUC 1.3 Itraconazole Cmax 1.0/1.2 AUC 1.0/1.2

tment C ea tment Cmax 0.97 AUC 0.95 Efavirenz Tr Cmax 0.99/0.99 AUC 0.99/0.98

Cmax 0.78 AUC 0.70 Rifampicin Cmax 0.89/0.80 AUC 0.87/0.76

Inducer Inhibitor

Strong Moderate Weak Weak Moderate Strong

0.01 0.1 1 10 100 Effect Size (log fold change) C

Predicted C max Predicted C max Cmax 1.6 Predicted AUC Predicted AUC Unity AUC 1.7 Ketoconazole HVs AML/advSM Cmax 1.4/1.5 AUC 1.5/1.6

Cmax 1.3 AUC 1.5 Fluconazole Cmax 1.3/1.4 AUC 1.41/5 on s ond iti Cmax 1.2 AUC 1.3 Itraconazole Cmax 1.2/1.2 AUC 1.2/1.2

tment C ea tment Cmax 0.97 AUC 0.96 Efavirenz Tr Cmax 0.98/0.99 AUC 0.97/0.98

Cmax 0.83 AUC 0.75 Rifampicin Cmax 0.85/0.85 AUC 0.78/0.80

Inducer Inhibitor

Strong Moderate Weak Weak Moderate Strong

0.01 0.1 1 10 100 Effect Size (log fold change) Figure 3 DMD Fast Forward. Published on November 8, 2017 as DOI: 10.1124/dmd.117.078006 This article has not been copyedited and formatted. The final version may differ from this version. Downloaded from dmd.aspetjournals.org at ASPET Journals on September 30, 2021

A Midostaurin single dose Midostaurin single dose (with rifampicin) B Midostaurin single dose Midostaurin single dose (with ketoconazole) Midostaurin twice daily Midostaurin twice daily (with rifampicin) Midostaurin twice daily Midostaurin twice daily (with ketoconazole) 700 350 (% ) (% ) 600 300

500 250

400 200 Enzyme Activity Enzyme Activity 300 150

200 100

100 50 3A4 Hepatic C YP 3A4 3A4 Hepatic C YP 3A4 0 0 0 67 134 201 268 335 402 469 536 603 670 0 67 134 201 268 335 402 469 536 603 670 Time (hours) Time (hours) Figure 4 DMD Fast Forward. Published on November 8, 2017 as DOI: 10.1124/dmd.117.078006 This article has not been copyedited and formatted. The final version may differ from this version. Downloaded from

A 60 Midazolam Midazolam 95th percentile 50 Midazolam 5th percentile dmd.aspetjournals.org

(ng /mL) Observed (without midostaurin) 40

30

20 at ASPET Journals on September 30, 2021 10 on Mi da zolam C on centrati

0 0 2 4 6 8 10 Time (hours) B 60 Midazolam + midostaurin (single dose) Midazolam 95th percentile 50 Midazolam 5th percentile (ng /mL) A2112 (day1) 40

30

20

10 on Mi da zolam C on centrati

0 0 2 4 6 8 10 Time (hours) C 60 Midazolam + midostaurin twice daily Midazolam 95th percentile 50 Midazolam 5th percentile

(ng /mL) A2112 (day 6) 40

30

20

10 on Mi da zolam C on centrati

0 120 122 124 126 128 130 Time (hours) Figure 5 Figure 6 C A CGP5421 Concentration (ng/mL) Midazolam Concentration (ng/mL) 1000 1200 1400 1600 200 400 600 800 10 12 14 16 18 20 0 2 4 6 8 0 4 4 5 5 5 5 658 656 654 652 650 648 646 51015203530455055650 585 520 455 390 325 260 195 130 65 0 Time(hours) Time(hours) This articlehasnotbeencopyeditedandformatted.Thefinalversionmaydifferfromthisversion. DMD FastForward.PublishedonNovember8,2017asDOI:10.1124/dmd.117.078006 Midazolam Midazolam + midostaurin + Midazolam D B CGP2221 Concentration (ng/mL) Midastaurin Concentration (ng/mL) 1000 1500 2000 2500 1000 1500 2000 2500 500 500 0 0 51015203530455055650 585 520 455 390 325 260 195 130 65 0 51015203530455055650 585 520 455 390 325 260 195 130 65 0

Time(hours) Time(hours)

at ASPET Journals on September 30, 2021 30, September on Journals ASPET at dmd.aspetjournals.org from Downloaded DMD Fast Forward. Published on November 8, 2017 as DOI: 10.1124/dmd.117.078006 This article has not been copyedited and formatted. The final version may differ from this version. Downloaded from dmd.aspetjournals.org at ASPET Journals on September 30, 2021

160 Rifampicin 140 Placebo Midostaurin 120 ± SD (ng/mL) 100

80

on on centrati 60

40 4βHC C 20

Mean 0 0 5 10 15 20 25 30 Time (days)

Midostaurin Time (days) 3 8 15 22 28 Predicted midazolam GM AUC ratio 0.84 0.60 0.55 0.54 0.54 with midostaurin Plasma 4 HC GM ratio relative to 1.2 1.4 1.8 1.8 1.9 baseline Rifampicin Time (days) 9 11 15 Plasma 4 HC GM ratio relative to 3.2 3.5 4.3 baseline Figure 7 PBPK modeling for midostaurin-CYP3A4 interaction

SUPPLEMENTAL DATA Authors: Helen Gu, Catherine Dutreix, Sam Rebello, Taoufik Ouatas, Lai Wang, Dung Yu Chun, Heidi J. Einolf, Handan He

Title: Simultaneous physiologically based pharmacokinetic (PBPK) modeling of parent and active metabolites to investigate complex CYP3A4 drug-drug interaction potential: a case example of midostaurin

Journal title: Drug Metabolism and Disposition

1 SUPPLEMENTARY MATERIALS AND METHODS

1.1 Materials The following chemicals and reagents were obtained from Sigma (St. Louis, MO): testosterone, 6β-hydroxytestosterone, midazolam, warfarin, β-NADPH, monobasic and dibasic potassium phosphate, dimethylsulfoxide (DMSO), rifampicin (RIF), and magnesium chloride. 1 ′ -

hydroxymidazolam was obtained from Corning Life Sciences - Discovery Labware (Woburn, MA) and [2H4]-1 ′ -hydroxymidazolam was obtained from Cerilliant (Round Rock, TX).

Phosphate-buffered saline was obtained from Gibco BRL (Invitrogen, Carlsbad, CA). Reverse transcriptase polymerase chain reaction (RT-PCR) probes, primers, and master mix; MagMAX- 96 total RNA isolation kit; RNase-free water; High-Capacity cDNA reverse transcription kit; and TaqMan universal PCR master mix were purchased from Applied Biosystems (Foster City, CA).

Human liver microsomes

Pooled human liver microsomes (n = 50 donors, mixed sex) were previously characterized by the vendors, as shown in Supplemental Table 1. PBPK modeling for midostaurin-CYP3A4 interaction

Supplemental Table 1. Pooled Human Liver Microsomes

Reversible Inhibition Time-Dependent Inhibition

Vendor XenoTech (Lenexa, KS) BD Biosciences

Lot 310198 82087

Cytochrome P450 content 0.425 0.29 (nmol/mg protein)

Cytochrome b5 content 0.546 0.79 (nmol/mg protein)

NADPH–cytochrome c reductase (nmol/min/mg 233 340 protein)

Human hepatocytes

Fresh primary human hepatocytes, lots FHU-L-070907 (liver 1, female), FHU-L-081407 (liver 2, female), and MHU-L-062009 (liver 3, male), were incubated on collagen-coated plates, InVitroGRO CP (plating) medium, InVitroGRO HI (incubation) medium, and Torpedo mix, obtained from In Vitro Technologies (Baltimore, MD).

1.2 In Vitro CYP3A4/5 Inhibition The potential of test compound to inhibit human CYP3A4/5 enzyme activity was assessed in pooled HLMs by testing the effect of increasing concentrations of midostaurin, CGP52421, or CGP62221 on the in vitro metabolism of two probe substrates with metabolisms known to be selective for CYP3A4/5 enzymes. CYP3A4/5 activity was assessed using the probes midazolam- 1′-hydroxylation or testosterone 6β-hydroxylation. Probe substrate concentrations used for these determinations were less than or equal to their reported Michaelis-Menten constant values. For reversible inhibition, the incubations (37°C, 10 min) were composed of (final concentrations) potassium phosphate buffer (100 mM, pH 7.4), nicotinamide adenine dinucleotide phosphate (NADPH; 1 mM), magnesium chloride (5 mM), HLM protein (0.1 mg protein/mL), probe substrate (5 μM midazolam or 25 μM testosterone), and varying concentrations of the test compounds (0-100 μM). After preincubation for 3 min, the reactions were initiated by addition of NADPH and terminated by addition of acetonitrile (200 μL). The organic solvent (1:1 acetonitrile:methanol [vol:vol]) content was less than 1%, with the PBPK modeling for midostaurin-CYP3A4 interaction

exception of midostaurin, which was increased to ≈ 5% due to the poor aqueous solubility of midostaurin in the HLM incubation mixture. Formation of probe substrate metabolites from the above samples, 1′-hydroxymidazolam and 6β-hydroxytestosterone, was determined by LC-

MS/MS after concentration and reconstitution of the samples in acetonitrile/water containing an internal standard. Values for 50% half maximal inhibitory concentration (IC50) for the inhibition of CYP enzyme were determined by visual inspection of the data (percentage of control CYP activity vs test compound concentration).

To investigate the possibility of TDI of CYP3A4/5, the test compounds (0-50 µM) were preincubated with HLM, using protein concentrations of 0.5 mg microsomal protein/mL. The concentrations of the test compounds were 1, 2, 5, 10, 25, and 50 µM for all experiments. After specified incubation times, aliquots (5 µL) of the preincubation mixture were transferred to 95- µL enzyme activity assay mixtures to determine the CYP3A4/5 activity remaining. The concentration of probe substrate in the enzyme activity assay was midazolam 20 μM. The enzyme activity reactions were incubated at 37°C for 6 min and terminated as above. Positive controls for CYP3A4/5 time-dependent inhibitor included troleandomycin. Sample preparation for analysis of probe substrate metabolite formation by LC-MS/MS was as described above. For data analysis, the inactivation parameters kinact (maximum inactivation rate) and KI (concentration

at 0.5kinact) were determined by plotting the natural log of the percentage of control activity remaining following incubations with increasing inhibitor concentration against time of

preincubation. The absolute value of the observed rate of inactivation (kobs) was then plotted against inhibitor concentration ([I]), and the data were analyzed by nonlinear regression using the equation:

kobs = (kinact [I])/(KI + [I]) (1)

1.3 In Vitro CYP3A4 Induction The potential for induction of CYP3A4 mRNA and enzyme activities by midostaurin, CGP52421, and CGP62221 was initially assessed in vitro using cryopreserved hepatocytes from three donors (Corning Life Sciences, Discovery Labware). The cells were treated with the test compounds (0.1-10 μM), positive controls RIF (0.1-10 μM), and vehicle control (0.1% DMSO) for 48 h. The PBPK modeling for midostaurin-CYP3A4 interaction

medium was changed with addition of fresh compounds or vehicle control 24 h after the first

treatment dose. Induction of mRNA was determined by real-time PCR using the comparative CT method, enzyme activity was measured in situ using midazolam as a CYP3A-selective probe substrate, and cell viability was assessed using the MTT (3-[4,5-dimethylthiazol-2-Yl]-2,5- diphenyltetrazolium bromide) assay after the treatment period. The method was essentially as

described in Flarakos et al (2016). Based on this initial assessment, values for EC50 and Emax for CYP3A4 mRNA induction were determined in three cryopreserved hepatocyte donors. The

induction parameters EC50 and Emax were determined by nonlinear regression of the mRNA induction data using a sigmoidal Emax model (2) or a sigmoidal 3-parameters model (3):

= ( × ) + (2) 𝑔𝑔 𝑔𝑔 𝑔𝑔 𝐸𝐸𝑓𝑓𝑓𝑓𝑓𝑓𝑓𝑓𝑓𝑓 = 𝐸𝐸𝑚𝑚𝑚𝑚𝑚𝑚 {1𝑋𝑋+ exp⁄�𝐸𝐸𝐸𝐸[ 50( 𝑋𝑋 � )/ ]} (3) 𝐸𝐸 𝐸𝐸𝐸𝐸𝐸𝐸𝐸𝐸𝐸𝐸 𝐸𝐸𝑚𝑚𝑚𝑚𝑚𝑚⁄ − 𝑋𝑋 − 𝐸𝐸𝐸𝐸50 𝑔𝑔 in which X is the inducer concentration in vitro and g represents the slope.

Indmax and IndC50 values used in the model were calibrated by values from positive control RIF within the Simcyp (v15, R1).

2 RESULTS

2.1 In Vitro CYP3A4/5 Inhibition The results of the in vitro CYP3A4/5 inhibition assessment are tabulated in Supplemental Table 2. Midostaurin, CGP52421, and CGP62221 were found to be reversible inhibitors of CYP3A4/5. PBPK modeling for midostaurin-CYP3A4 interaction

Supplemental Table 2. Effect of Midostaurin, CGP52421, and CGP62221 on CYP3A4/5 Probe Substrate Metabolism a CYP Probe Reaction Total Ki, IC50/2 (unbound; µM) Enzyme (reference) Midostaurin CGP52421 CGP62221 Midazolam 1′- 0.75 (0.25) 0.75 (0.44) 0.4 (0.25) hydroxylation CYP3A4/5 Testosterone 6β- hydroxylation > 100 1 0.5

a Unbound values corrected for microsomal protein binding were based on the following predicted values (ADMET Predictor, V 6.5): fumic was 0.38 for midostaurin, 0.59 for CGP52421, and 0.62 for CGP62221.

TDI of CYP3A4/5 was observed for all three test articles, and the inhibition was concentration

dependent. The inactivation parameters KI and kinact associated with this TDI are shown in Supplemental Table 3.

Supplemental Table 3. CYP3A4/5 TDI Results −1 Test Compound Human Probe Reaction Total KI (µM) kinact (min ) Enzyme (unbound; µM)a Troleandomycinb 0.233 ± 0.0206 0.0663 ± 0.00114 1.02 ± 0.134 Midostaurin 0.0466 ± 0.00114 (0.0908 ± 0.0119) Midazolam 1′- CYP3A4/5 1.97 ± 0.230 CGP52421 hydroxylation 0.0502 ± 0.00135 (0.453 ± 0.0529) 2.01 ± 0.321 CGP62221 0.0763 ± 0.00281 (0.503 ± 0.0803)

a Unbound values corrected for microsomal protein binding were based on the following predicted values (ADMET Predictor, V 6.5): fumic was 0.089 for midostaurin, 0.23 for CGP52421, and 0.25 for CGP62221. b Positive control. Considering the complexities associated with predictions of time- and concentration-dependent effects, PBPK modeling using a bottom-up approach (ie, based solely on in vitro results) is not recommended (FDA, 2012; EMA, 2012). In a general practice, both unbound Ki and KI should be

used in the model. However, in our top-down approach, in vitro study-derived parameters (KI

[total] and kinact for TDI) were entered to best describe the clinically observed concentration-time profiles after multiple dosing. In order to address the discrepancy between the use of inhibition parameters either corrected or uncorrected for microsomal protein binding, a parameter PBPK modeling for midostaurin-CYP3A4 interaction sensitivity analysis of midostaurin, CGP52421, and CGP62221 fumic value for the KI value (range of the unbound value to total value) on the predicted midazolam AUC ratio was conducted.

Supplemental Figure 1. Sensitivity Analysis for the Impact of fumic Values for the KI on the Predicted Midazolam AUC Ratio Dosing regimen: Midazolam 5 mg on day 28 + midostaurin 50 mg BID on days 1-28 in HV

(A) Varying midostaurin fumic from 0.089 to 1 for the KI value resulted in little change in the predicted midazolam AUC ratio (12% decrease) in the absence and presence of midostaurin

(B) Varying CGP52421 fumic from 0.23 to 1 for the KI value resulted in little change in the predicted midazolam AUC ratio (8% decrease) in the absence and presence of midostaurin PBPK modeling for midostaurin-CYP3A4 interaction

(C) Varying CGP62221 fumic from 0.25 to 1 for the KI value resulted in little change in the predicted midazolam AUC ratio (27% decrease) in the absence and presence of midostaurin

The results of this parameter sensitivity analysis indicated that CYP3A4/5 DDI was not highly sensitive to the midostaurin and its metabolites (as perpetrators – inducers) fumic for the KI value evaluated.

2.2 In Vitro CYP3A4 Induction The results of the preliminary in vitro CYP induction study in three donors of human hepatocytes are shown in Supplemental Table 4. Dose-dependent increases in CYP3A4 mRNA levels with PBPK modeling for midostaurin-CYP3A4 interaction the test article treatment (up to 25 µM) were observed in the three donors. Induction of CYP3A4 mRNA exceeded two-fold in the three donors; therefore, a follow-up in vitro induction study to determine the induction parameters Emax and EC50 for mRNA was performed in one hepatocyte donor. Induction of CYP3A4/5 mRNA with little change in its enzyme activity was observed and was consistent with the time-dependent inhibition properties of midostaurin, CGP52421, and CGP62221.

Supplemental Table 4. Fold Change in CYP3A4 mRNA and Activity in Human Hepatocytes Treated with Midostaurin and Its Metabolites or Positive Control Inducer, Rifampicin Donor 1 Donor 2 Donor 3 Treatment mRNA Activity mRNA Activity mRNA Activity Midostaurin 0.1 µM NA NA NA NA 0.233 1.05

Midostaurin 0.25 µM NA NA NA NA 0.879 1.93

Midostaurin 0.5 µM 9.39 1.51 4.35 1.15 3.35 2.65

Midostaurin 1 µM 25.8 1.97 8.58 0.632 9.35 3.64

Midostaurin 2.5 µM NA NA NA NA 12.2 3.38

Midostaurin 5 µM 40.1 1.64 9.03 0.896 7.35 2.69

Midostaurin 7.5 µM NA NA NA NA 6.37 2.69

Midostaurin 10 µM 25.8 1.75 7.51 1.06 4.81 2.88

Midostaurin 25 µM 24.7 1.61 6.67 1.02 NA NA

Emax 12.5 ± 0.32

EC50 0.696 ± 0.025

CGP52421 0.1 µM NA NA NA NA 0.11 0.876

CGP52421 0.25 µM NA NA NA NA 0.289 1.07

CGP52421 0.5 µM 9.36 1.08 3.47 1.50 1.28 1.54

CGP52421 1 µM 24.4 1.54 7.17 1.43 2.52 2.62

CGP52421 2.5 µM NA NA NA NA 14.0 4.00

CGP52421 5 µM 56.3 1.81 10.5 1.25 14.4 4.38

CGP52421 10 µM 35.9 1.57 9.34 1.13 15.2 3.95

CGP52421 25 µM 43.6 1.76 8.13 1.33 NA NA PBPK modeling for midostaurin-CYP3A4 interaction

Emax 14.9 ± 0.48

EC50 1.41 ± 0.12 CGP62221 0.1 µM NA NA NA NA 0.39 1.24 CGP62221 0.25 µM NA NA NA NA 1.63 1.94 CGP62221 0.5 µM 7.69 2.00 4.97 1.45 4.38 2.83 CGP62221 1 µM 21.9 2.18 6.78 1.35 11.4 4.53 CGP62221 2.5 µM NA NA NA NA 19.9 5.90 CGP62221 5 µM 47.9 3.07 8.54 1.35 15.0 3.69 CGP62221 10 µM 42.8 2.40 8.88 1.19 14.7 2.67

Emax 16.5 ± 1.4

EC50 0.710 ± 0.13 Rifampin 0.1 µM NA NA NA NA 0.817 1.93 Rifampin 0.25 µM NA NA NA NA 1.50 3.31 Rifampin 0.5 µM 20.6 5.49 5.69 7.84 2.87 4.14 Rifampin 1 µM 55.1 8.98 6.47 9.44 5.76 12.4 Rifampin 2.5 µM NA NA NA NA 6.88 12.5 Rifampin 5 µM 62.2 12.1 6.92 12.1 14.2 20.2 Rifampin 7.5 µM NA NA NA NA 10.9 19.7 Rifampin 10 µM 54.8 13.9 6.62 13.5 16.6 22.9 Rifampin 25 µM 42.2 14.6 6.65 12.1 19.6 21.9

Emax 22.2 ± 2.6

EC50 4.21 ± 1.4 NA, not applicable.

2.3 PBPK- and Pop PK–Simulated PK Parameters in Patients With AML or advSM (authors are grateful to Dr. Zisowsky for providing Pop PK data) The PBPK- and Pop PK–simulated PK parameters of midostaurin and its metabolites in patients with AML or advSM can be found in Supplemental Tables 5 and 6, respectively. Simulated

AUC and Cmax values for midostaurin and its metabolites were within ≤ 2-fold of the Pop PK– simulated values. PBPK modeling for midostaurin-CYP3A4 interaction

Supplemental Table 5. Summary of PBPK- and Pop PK–Simulated Midostaurin, CGP52421, and CGP62221 AUC and Cmax in Patients With AML After Midostaurin 50 mg bid Pop PK Pop PK PBPK Simulated PBPK Simulated Compound Day Simulated Simulated Mean ± SD Mean ± SD Mean Mean

AUC0-24h (h•ng/mL) Cmax (ng/mL) Midostaurin 1 37,893 33,603 ± 25,083 2535 3553 ± 2436 28 43,110 43,739 ± 25,075 2685 4025 ± 1963 CGP52421 1 7372 7098 ± 8424 452 425 ± 498 28 131,973 115,855 ± 81,541 5558 4908 ± 3444 CGP62221 1 13,917 11,008 ± 10,187 945 661 ± 620 28 36,295 44,817 ± 23,412 1548 2049 ± 994

Supplemental Table 6. Summary of PBPK- and Pop PK–Simulated Midostaurin, CGP52421, and CGP62221 AUC and Cmax in Patients With advSM After Midostaurin 100 mg bid Pop PK Pop PK PBPK Simulated PBPK Simulated Compound Day Simulated Simulated Mean ± SD Mean ± SD Mean Mean

AUC0-24h (h•ng/mL) Cmax (ng/mL) Midostaurin 1 58,766 ± 20,118 54,153 ± 44,378 3764 ± 1240 4617 ± 3447 28 50,341 ± 21,388 64,677 ± 60,765 3370 ± 1141 4786 ± 3482 CGP52421 1 8320 ± 2370 7529 ± 8035 518 ± 142 463 ± 496 28 66,456 ± 11,4437 47,962 ± 32,483 2810 ± 604 2110 ± 1350 CGP62221 1 27208 ± 7185 16,180 ± 15,905 1770 ± 413 983 ± 1016 28 57,682 ± 19,280 67,605 ± 59,613 2516 ± 804 3041 ± 2503

2.4 Representative Actual and PBPK-Simulated Clinical PK Parameters in HV The PBPK model was verified by simulating several clinical trials in which single-agent PK was available (shown in Figure 1). The representative tabulated simulated and actual PK parameters for midostaurin, CGP52421, and CGP62221 following a single dose of midostaurin 50 mg and 50 mg bid can be found in Supplemental Tables 7 and 8. Overall, the PKs of midostaurin, CGP52421, and CGP62221 were predicted reasonably well. The predicted GM values, AUC, and Cmax were within ≤ 2-fold of the observed values. PBPK modeling for midostaurin-CYP3A4 interaction

Supplemental Table 7. Representative Actual and Simulated Clinical PK Parameters for Midostaurin, CGP52421, and CGP62221 After a Single Dose of Midostaurin 50 mg Parameter Midostaurin CGP52421 CGP62221 Cmax (ng/mL) Predicted 1253 (80) 198 (125) 526 (82) GM (%CV) Observeda 1328 (20.6) 222 (23.7) 538 (11.5) PEb −6% −11% −2% AUC0-120h Predicted 10109 (64) 10726 (89) 16890 (78) (h•ng/mL) Observeda 12588 (32.6) 9984 (34.3) 20195 (22.51) GM (%CV) PEb −20% +7% −16% a Observed data: A2111. b Prediction error = (simulated − observed)/observed × 100.

PBPK modeling for midostaurin-CYP3A4 interaction

Supplemental Table 8. Actual and Simulated Clinical PK Parameters for Midostaurin, CGP52421, and CGP62221 Following Multiple Oral Doses of Midostaurin 50 mg Parameter Midostaurin CGP52421 CGP62221 Day −1 Predicted 1252 (80) 198 (125) 525 (82) a Cmax (ng/mL) Observed 1206 (19.3) 209 (20.0) 532 (10.0) GM (%CV) PEb +4% −5% −1% AUC0-12h Predicted 5704 (80) 1699 (129) 4624 (85) (h•ng/mL) Observeda 8133 (16.4) 1882 (19.8) 4946 (13.2) GM (%CV) PEb −30% −10% −7% Day −7 Predicted 2041 (62) 979 (77) 1448 (72) a Cmax (ng/mL) Observed 1612 (27.9) 1177 (19.2) 1668 (24.3) GM (%CV) PEb +27% −17% −13% AUC0-12h Predicted 8390 (67) 10,528 (74) 14,543 (76) (h•ng/mL) Observeda 13,103 (29) 13,152 (21.2) 18,384 (25.1) GM (%CV) PEb −36% −20% −21% a Observed data: A2116. b Prediction error = (simulated − observed)/observed × 100. PBPK modeling for midostaurin-CYP3A4 interaction

Supplemental Table 9.Subjects in Trials Used for Model Development and Validation Study Subjects Groups Purpose of Study Trial Identifiera and/or Reference A2108 Healthy Arm 1: single dose of midostaurin 50 mg (FMI gelatin capsule) (n = 18) Biopharmaceutics volunteers Arm 2: single dose of midostaurin 50 mg (CSF gelatin capsule) (n = 18) (testing bioavailability of aged 25-50 y Arm 3: single dose of midostaurin 50 mg (pediatric oral solution) (n = 18) various formulations) A2109 Healthy Arm 1: ketoconazole 400 mg qd on days 1-10, single dose of midostaurin 50 DDIs (effect of EudraCT volunteers mg on day 6 (n = 27) ketoconazole, a strong 2008-003038-39 aged 18-55 y Arm 2: placebo qd on days 1-10, single dose of midostaurin 50 mg on day 6 (n CYP3A4 inhibitor, on (Dutreix 2013) = 20) midostaurin PK) A2110 Healthy Arm 1: rifampicin 600 mg qd on days 1-14, single dose of midostaurin 50 mg DDIs (effect of EudraCT volunteers on day 9 (n = 25) rifampicin, a potent 2009-009895-11 aged 18-55 y Arm 2: placebo qd on days 1-14, single dose of midostaurin 50 mg on day 9 (n CYP3A4 inducer, on (Dutreix 2013; Dutreix = 22) midostaurin PK) 2014) A2111 Healthy Arm 1: midostaurin 50 mg (capsule) with a standard meal (n = 12) Biopharmaceutics Approved by the State volunteers Arm 2: midostaurin 25 mL (oral solution) with a standard meal (n = 12) (bioavailability:food Office of Health and aged 18-55 y Arm 3: midostaurin 50 mg with a high-fat meal (n = 12) effect) Social Affairs Ethics Arm 4: midostaurin 50 mg while fasting (n = 12) Committee of the Land Berlin (Novartis data on file) A2112 Healthy Midazolam 4 mg with standard breakfast on day 1; midostaurin 100 mg + DDIs (effect of EudraCT volunteers midazolam 4 mg with standard breakfast on day 3; midostaurin 50 mg bid with midostaurin on the PK of 2009-009870-29 aged 18-55 y standard breakfast on days 4-6; midazolam 4 mg with standard breakfast on midazolam, a CYP3A4 (Dutreix 2013) day 8 (n = 20) probe) A2113 Healthy Arm 1: midostaurin 75 mg bid on days 1 and 2, single dose of placebo on day Cardiac safety del Corral 2012 volunteers 3 (n = 80) aged 18-45 y Arm 2: placebo bid on days 1 and 2, single dose of placebo on day 3 (n = 68) Arm 3: placebo bid on days 1 and 2, single dose of moxifloxacin 400 mg on day 3 (n = 44) A2116 Healthy Midostaurin 50 mg bid on days 1-6 and single dose of 50 mg on day 7 (N = Impact of liver NCT01429337 volunteers 42) dysfunctionb Ongoing trial aged 18-70 y Arms: normal liver function; mild, moderate, and severe hepatic impairment (ClinicalTrials.gov) bid, twice daily; CSF, clinical service form; CYP3A4, cytochrome P450 3A4; DDI, drug-drug interaction; FMI, final market image; qd, once daily; PK, pharmacokinetics. a Trial registration and/or approval information is provided where available. b Control arm with normal liver function was reported in the current study.

PBPK modeling for midostaurin-CYP3A4 interaction

Supplemental Table 10. PBPK Model Input Parameters for Midazolam (Sim-Midazolam) Parameter Value Physical chemistry and blood binding Molecular weight (g/mol) 325.8 logP 3.53 Compound type Ampholyte pKa1 10.95 pKa2 6.2 B/P 0.603 fup 0.032 Absorption Model used First-order absorption Fa 1 Lag time (h) 0 −1 ka (h ) 3 fugut 1 Qgut (L/h) 14.02 Peff, man (10−4 cm/s) 6.045 Caco-2 permeability (106 cm/s) (scalar) 213 (0.29) Distribution Model used Minimal PBPK −1 kin (h ) 0.2 −1 kout (h ) 0.25 Vsac (L/kg) 0.23 Vss (L/kg) 0.88 Kp (scalar) 1 Elimination Model used Enzyme kinetics, recombinant CYP CYP3A4 Vmax (pmol/min/pmol of CYP) 5.23 (1-OH pathway), 5.2 (4-OH pathway) CYP3A4 Kmu (µM) 2.16 (1-OH pathway), 31.8 (4-OH pathway) CYP3A5 Vmax (pmol/min/pmol of CYP) 19.7 (1-OH pathway), 4.03 (4-OH pathway) CYP3A5 Kmu (µM) 4.16 (1-OH pathway), 34.8 (4-OH pathway) UGT1A4 Vmax (pmol/min/pmol of UGT) 445 UGT1A4 Kmu (µM) 40.3 Active uptake into hepatocytes 1 CLR (L/h) 0.085 B/P, blood-to-plasma ratio; Caco-2, continuous heterogeneous human epithelial colorectal adenocarcinoma cell line; CLR, renal clearance; Fa, fraction of dose absorbed; fugut, fraction of unbound drug in the gut; fup, fraction of unbound drug in plasma; ka, absorption rate constant; Kin, zero-order rate constant for the formation of a physiological response variable; Kmu, unbound Michaelis-Menten constant; Kout, first-order rate constant for the degradation of a physiological response variable; Kp, tissue:plasma partition coefficient; logP, log of the partition coefficient; PBPK, physiologically based pharmacokinetics; Peff, man, effective permeability in man; pKa, acid dissociation constant; Qgut, nominal flow through the gut; Vmax, maximum velocity; Vsac, volume of distribution for the single adjustable compartment; Vss, volume of distribution at steady state.

14

PBPK modeling for midostaurin-CYP3A4 interaction

Supplemental Table 11. PBPK Model Input Parameters for Ketoconazole (Sim-Ketoconazole 400 mg qd) Parameter Value Physical chemistry and blood binding Molecular weight (g/mol) 531.4 logP 4.04 Compound type Diprotic base pKa1 2.94 pKa2 6.51 B/P 0.62 fup 0.029 Absorption Model used First-order absorption Fa 1 Lag time (h) 0 −1 ka (h ) 0.78 fugut 0.06 Qgut (L/h) 13.23 Peff, man (10−4 cm/s) 4.947 PSA (Å2) 69.06 HBD 0 Distribution Model used Minimal PBPK Vss (L/kg) 0.345 Elimination Model used In vivo clearance CLpo (CV %) (L/h) 7.4 (40) Active uptake into hepatocytes 2.07 CLR (L/h) 0.147 Interaction CYP2C8 Ki (fumic) (µM) 2.5 (0.87) CYP2C9 Ki (fuinc) (µM) 10 (0.95) CYP3A4 Ki (fumic) (µM) 0.015 (0.97) CYP3A5 Ki (fumic) (µM) 0.109 (0.96) B/P, blood-to-plasma ratio; CLpo, oral clearance; CLR, renal clearance; CV, coefficient of variation; CYP, cytochrome; Fa, fraction of dose absorbed; fugut, fraction of unbound drug in the gut; fuinc, fraction of unbound drug in the in vitro incubation; fumic, fraction of unbound drug in microsomes; fup, fraction of unbound drug in plasma; HBD, hydrogen bonding donor; ka, absorption rate constant; Ki, dissociation constant for an inhibitor; logP, log of the partition coefficient; PBPK, physiologically based pharmacokinetics; pKa, acid dissociation constant; Peff, man, effective permeability in man; PSA, polar surface area; Qgut, nominal flow through the gut; Vss, volume of distribution at steady state.

15

PBPK modeling for midostaurin-CYP3A4 interaction

Supplemental Table 12. PBPK Model Input Parameters for Itraconazole and Its Metabolite Itraconazole (SV-Itraconazole-Fed Capsule) Parameter Value Physical chemistry and blood binding Molecular weight (g/mol) 705.6 logP 4.47 Compound type Monoprotic base pKa 4.28 B/P 0.58 fup 0.016 Absorption Model used First-order absorption Fa 0.59 Lag time (h) 0 −1 ka (h ) 0.6 fugut 0.016 Qgut (L/h) 15.65 Peff, man (10−4 cm/s) 9.85 MDCK II (scalar) (106 cm/s) 57.1 (1.34) Distribution Model used Minimal PBPK Vss (L/kg) 2.52 Kp (scalar) 0.3 Elimination Model used Enzyme kinetics, recombinant CYP CYP1A2 CLint (µL/min/pmol CYP) 1 CYP3A4 Vmax (pmol/min/pmol of CYP) 0.065 CYP3A4 Kmu (µM) 0.0039 Active uptake into hepatocytes 1 CLR (L/h) 0 Interaction CYP3A4 Ki (fumic) (μM) 0.0013 (1) Metabolite of Itraconazole (SV-OH-Itraconazole) Parameter Value Physical chemistry and blood binding Molecular weight (g/mol) 721.7 logP 4.47 Compound type Monoprotic base pKa 4.28 B/P 0.58 fup 0.016 Distribution Model used Minimal PBPK Vss (L/kg) 1.035 Kp (scalar) 0.12

16

PBPK modeling for midostaurin-CYP3A4 interaction

Elimination Model used Enzyme kinetics, recombinant CYP CYP3A4 Vmax (pmol/min/pmol of CYP) 0.13 CYP3A4 Kmu (µM) 0.027 Active uptake into hepatocytes 1 CLR (L/h) 1.39 Interaction CYP3A4 Ki (fumic) (μM) 0.0023 (1) B/P, blood-to-plasma ratio; CLint, intrinsic clearance; CLR, renal clearance; CYP, cytochrome; Fa, fraction of dose absorbed; fugut, fraction of unbound drug in the gut; fumic, fraction of unbound drug in microsomes; fup, fraction of unbound drug in plasma; ka, absorption rate constant; Ki, dissociation constant for an inhibitor; Kmu, unbound Michaelis-Menten constant; Kp, tissue:plasma partition coefficient; logP, log of the partition coefficient; MDCK, Madin-Darby canine kidney cell line; PBPK, physiologically based pharmacokinetics; Peff, man, effective permeability in man; pKa, acid dissociation constant; Qgut, nominal flow through the gut;Vmax, maximum velocity; Vss, volume of distribution at steady state.

17

PBPK modeling for midostaurin-CYP3A4 interaction

Supplemental Table 13. PBPK Model Input Parameters for Fluconazole (SV-Fluconazole) Parameter Value Physical chemistry and blood binding Molecular weight (g/mol) 306.3 logP 0.2 Compound type Monoprotic base pKa 1.76 B/P 1 fup 0.89 Absorption Model used First-order absorption Fa 0.986 Lag time (h) 0 −1 ka (h ) 1.76 fugut 0.89 Qgut (L/h) 12.60 Peff, man (10−4 cm/s) 4.266 Caco-2 permeability (scalar) (106 cm/s) 29.8 (0.885) PSA (Å2) 69.06 HBD 0 Distribution Model used Minimal PBPK Vss (L/kg) 0.748 Elimination Model used In vivo clearance CLiv (CV %) (L/h) 1.01 (24) Active uptake into hepatocytes 1 CLR (L/h) 0.7 Interaction CYP2C9 Ki (fuinc) (µM) 7.92 (1) CYP2C19 Ki (fuinc) (µM) 2 (1) CYP3A4 Ki (fumic) (µM) 10.7 (1) CYP3A5 Ki (fumic) (µM) 84.6 (1) B/P, blood-to-plasma ratio; Caco-2, continuous heterogeneous human epithelial colorectal adenocarcinoma cell line; CLiv, hepatic blood

clearance; CV, coefficient of variation; CYP, cytochrome; Fa, fraction of dose absorbed; fugut, fraction of unbound drug in the gut; fumic, fraction

of unbound drug in microsomes; fup, fraction of unbound drug in plasma; HBD, hydrogen bonding donor; ka, absorption rate constant; Ki, dissociation constant for an inhibitor; logP, log of the partition coefficient; PBPK, physiologically based pharmacokinetics; Peff, man, effective permeability in man; pKa, acid dissociation constant; PSA, polar surface area; Qgut, nominal flow through the gut; Vss, volume of distribution at steady state.

18

PBPK modeling for midostaurin-CYP3A4 interaction

Supplemental Table 14. PBPK Model Input Parameters for Rifampicin (SV-Rifampicin-MD) Parameter Value Physical chemistry and blood binding Molecular weight (g/mol) 823 logP 4.01 Compound type Ampholyte pKa1 1.7 pKa2 7.9 B/P 0.9 fup 0.116 Absorption Model used First-order absorption Fa 0.915 Lag time (h) 0 −1 ka (h ) 0.885 fugut 1 Qgut (L/h) 9.42 Peff, man (10−4 cm/s) 2.15 Caco-2 permeability (scalar) (106 cm/s) 15 (1) Distribution Model used Minimal PBPK Vss (CV %) (L/kg) 0.42 (50) Elimination Model used In vivo clearance CLiv (L/h) 8.7 Active uptake into hepatocytes 1 CLR (L/h) 1.26 Interaction CYP2C8 Ki (fumic) (μM) 24.5 (1) CYP3A4 Ki (fumic) (μM) 15 (1) CYP3A4 Indmax (fold) 16 CYP3A4 IndC50 (fuinc) (μM) 0.32 (1) CYP3A5 Indmax (fold) 16 CYP3A5 IndC50 (fuinc) (μM) 0.32 (1) B/P, blood-to-plasma ratio; Caco-2, continuous heterogeneous human epithelial colorectal adenocarcinoma cell line; CLiv, hepatic blood clearance; CLR, renal clearance; CV, coefficient of variation; CYP, cytochrome; Fa, fraction of dose absorbed; fugut, fraction of unbound drug in

the gut; fuinc, fraction of unbound drug; fumic, fraction of unbound drug in microsomes; fup, fraction of unbound drug in plasma; IndC50, calibrated concentration of inducer at half-Indmax; Indmax, maximum induction rate; ka, absorption rate constant; Ki, dissociation constant for an inhibitor; logP, log of the partition coefficient; PBPK, physiologically based pharmacokinetics; Peff, man, effective permeability in man; pKa, acid dissociation constant; Qgut, nominal flow through the gut; Vss, volume of distribution at steady state.

19

PBPK modeling for midostaurin-CYP3A4 interaction

Supplemental Table 15. PBPK Model Input Parameters for Efavirenz (SV-Efavirenz) Parameter Value Physical chemistry and blood binding Molecular weight (g/mol) 315.68 logP 4.02 Compound type Monoprotic acid pKa 10.2 B/P 0.74 fup 0.029 Absorption Model used First-order absorption Fa (CV %) 0.67 (15) Lag time (h) 0 −1 ka (CV %) (h ) 0.41 (15) fugut 0.005 Qgut (L/h) 13.78 Peff, man (10−4 cm/s) 5.680 PSA, Å2 38.33 HBD 1 Distribution Model used Minimal PBPK −1 kin (h ) 0.29 −1 kout (h ) 0.09 Vsac (L/kg) 1.1 Vss (CV %) (L/kg) 2.256 Kp (scalar) 0.155 Elimination Model used Enzyme kinetics, recombinant CYP CYP1A2 CLint (µL/min/pmol CYP) 0.03 CYP2B6 CLint (µL/min/pmol CYP) 1.36 CYP2A6 CLint (µL/min/pmol CYP) 0.47 CYP3A4 CLint (µL/min/pmol CYP) 0.012 Additional HLM CLint (µL/min/mg protein) 0.694 Active uptake into hepatocytes 1 CLR (L/h) 0 Interaction CYP2B6 Indmax (fold) 6.2 CYP2B6 IndC50 (fuinc) (µM) 1.2 (0.15) CYP3A4 Indmax (fold) 9.9 CYP3A4 IndC50 (fuinc) (µM) 3.8 (0.15) B/P, blood-to-plasma ratio; CLint, intrinsic clearance; CLR, renal clearance; CV, coefficient of variation; CYP, cytochrome; HLM, human liver microsome; IndC50, calibrated concentration of inducer at half-Indmax; Fa, fraction of dose absorbed; fugut, fraction of unbound drug in the gut; fuinc, fraction of unbound drug; fup, fraction of unbound drug in plasma; HBD, hydrogen bonding donor; Indmax, maximum induction rate; ka, absorption rate constant; Kin, zero-order rate constant for the formation of a physiological response variable; Kp, tissue:plasma partition coefficient; Kout, first-order rate constant for the degradation of a physiological response variable; logP, log of the partition coefficient; PBPK, physiologically based pharmacokinetics; Peff, man, effective permeability in man; pKa, acid dissociation constant; PSA, polar surface area; Qgut, nominal flow through the gut; Vsac, volume of distribution for the single adjustable compartment; Vss, volume of distribution at steady state.

20

PBPK modeling for midostaurin-CYP3A4 interaction

3 Supplemental References ClinicalTrials.gov. Pharmacokinetics and safety of midostaurin in subjects with impaired hepatic

function and subjects with normal hepatic function.

https://clinicaltrials.gov/ct2/show/NCT01429337. Accessed August 8, 2017.

del Corral A, Dutreix C, Huntsman-Labed A, Lorenzo S, Morganroth J, Harrell R, and Wang Y

(2012) Midostaurin does not prolong cardiac repolarization defined in a thorough electrocardiogram trial in healthy volunteers. Cancer Chemother Pharmacol 69: 1255–1264.

Dutreix C, Munarini F, Lorenzo S, Roesel J, and Wang Y (2013) Investigation into CYP3A4-

mediated drug-drug interactions on midostaurin in healthy volunteers. Cancer Chemother

Pharmacol 72: 1223–1234.

Dutreix C, Lorenzo S, and Wang Y (2014) Comparison of two endogenous biomarkers of

CYP3A4 activity in a drug-drug interaction study between midostaurin and rifampicin. Eur J

Clin Pharmacol 70: 915–920.

Flarakos J, Du Y, Bedman T, Al-Share Q, Jordaan P, Chandra P, Albrecht D, Wang L, Gu H,

Einolf HJ, Huskey SE, Mangold JB (2016). Disposition and metabolism of [(14)C]

Sacubitril/Valsartan (formerly LCZ696) an angiotensin receptor neprilysin inhibitor, in healthy subjects. Xenobiotica. 2016;46:986-1000.

21