throbber
ORIGINAL ARTICLE
`
`Effect of CYP3A perpetrators on ibrutinib exposure in
`healthy participants
`Jan de Jong1,, Donna Skee2, Joe Murphy2, Juthamas Sukbuntherng3, Peter Hellemans4, Johan Smit4,
`Ronald de Vries4, Juhui James Jiao2, Jan Snoeys4 & Erik Mannaert4
`1Janssen Research & Development, San Diego, California
`2Janssen Research & Development, Raritan, New Jersey
`3Pharmacyclics Inc., Sunnyvale, California
`4Janssen Research & Development, Beerse, Belgium
`
`Keywords
`Bioavailability, CYP3A perpetrators, grapefruit
`juice, ibrutinib, ketoconazole, rifampin
`
`Correspondence
`Jan de Jong, 3210 Merryfield Row, San
`Diego, CA 92121. Tel: 1 (858) 320-3480;
`E-mail: jdejong1@its.jnj.com
`
`Funding Information
`This study was supported by funding from
`Janssen Research & Development, LLC.
`
`Received: 15 May 2015; Accepted: 24 May
`2015
`
`Pharma Res Per, 3(4), 2015, e00156,
`doi: 10.1002/prp2.156
`
`doi: 10.1002/prp2.156
`
`Previous Presentation: American Society for
`Clinical Pharmacology and Therapeutics
`(ASCPT), March 18–24, 2014.
`
`Abstract
`
`Ibrutinib (PCI-32765), a potent covalent inhibitor of Bruton’s tyrosine kinase,
`has shown efficacy against a variety of B-cell malignancies. Given the prominent
`role of CYP3A in ibrutinib metabolism, effect of coadministration of CYP3A
`perpetrators with ibrutinib was evaluated in healthy adults. Ibrutinib (120 mg
`[Study 1, fasted], 560 mg [studies 2 (fasted), and 3 (nonfasted)]) was given
`alone and with ketoconazole [Study 1; 400 mg q.d.], rifampin [Study 2;
`600 mg q.d.], and grapefruit juice [GFJ, Study 3]. Lower doses of ibrutinib
`were used together with CYP3A inhibitors [Study 1: 40 mg; Study 3: 140 mg],
`as safety precaution. Under fasted condition, ketoconazole increased ibrutinib
`dose-normalized (DN) exposure [DN-AUClast: 24-fold; DN-Cmax: 29-fold],
`rifampin decreased ibrutinib exposure [Cmax: 13-fold; AUClast: 10-fold]. Under
`nonfasted condition, GFJ caused a moderate increase [DN-Cmax: 3.5-fold; DN-
`AUC: 2.2-fold], most likely through inhibition of intestinal CYP3A. Half-life
`was not affected by CYP perpetrators indicating the interaction was mainly on
`first-pass extraction. All treatments were well-tolerated.
`
`Abbreviations
`AE, adverse events; BCR, B-cell antigen receptor; BTK, Bruton’s tyrosine kinase;
`DDI, drug–drug interaction; GFJ, grapefruit juice; LC-MS/MS, liquid chromatogra-
`phy-tandem mass spectroscopy; PBPK, physiologically based pharmacokinetic.
`
`Introduction
`
`B-cell antigen receptor (BCR) signaling is implicated as a
`pivotal pathway in tumorigenesis in majority of B-cell
`malignancies. Antigen stimulation of normal B cells trig-
`gers dimerization of BCR initiating a downstream signal-
`ing kinase cascade, which in turn regulates multiple
`cellular processes, including proliferation, differentiation,
`apoptosis, and survival (Fuentes-Panana et al. 2004; Ad-
`vani et al. 2013). Bruton’s tyrosine kinase (BTK), a criti-
`cal terminal kinase enzyme in the BCR signaling pathway,
`is a promising target
`for therapeutic intervention in
`
`human malignancies. This downstream signal transduc-
`tion protein plays a key role in the activation of pathways
`necessary for B-cell trafficking, chemotaxis, and adhesion,
`and has also been implicated in initiation, survival, and
`progression of mature B-cell
`lymphoproliferative disor-
`ders (Kuppers 2005).

`, PCI–32765), an orally active,
`Ibrutinib (Imbruvica
`BTK-targeting inhibitor, has been recently approved for
`the treatment of patients with chronic lymphocytic leuke-
`mia and mantle cell lymphoma who have received at least
`one prior therapy; Ibrutinib being the first covalent inhib-
`itor of BTK to be advanced into human clinical trials. It
`
`ª 2015 Janssen Research and Development, LLC. Pharmacology Research & Perspectives published by John Wiley & Sons Ltd,
`British Pharmacological Society and American Society for Pharmacology and Experimental Therapeutics.
`This is an open access article under the terms of the Creative Commons Attribution-NonCommercial-NoDerivs License,
`which permits use and distribution in any medium, provided the original work is properly cited, the use is non-commercial and
`no modifications or adaptations are made.
`
`2015 | Vol. 3 | Iss. 4 | e00156
`Page 1
`
`SANDOZ INC.
`
`IPR2023-00478
`
`Ex. 1036, p. 1 of 11
`
`

`

`CYP3A Perpetrators and Ibrutinib Exposure
`
`Jan de Jong et al.
`
`forms a stable covalent bond with cysteine-481 on the
`active site of BTK and irreversibly inhibits BTK phos-
`phorylation on Tyr223, impairing BCR signaling, and dis-
`rupting the proliferation and survival of malignant B-cells
`Ibrutinib is
`(IC50: 0.39 nM) (Honigberg et al. 2010).
`almost
`exclusively metabolized by cytochrome P450
`(CYP) CYP3A. Absolute oral bioavailability (F) is low,
`ranging from 3.9% in the fasted state to 8.4% following a
`standard breakfast without grapefruit
`juice (GFJ) and
`15.9% with GFJ (de Vries et al. 2015). A major metabo-
`lite of ibrutinib, PCI-45227, is a dihydrodiol metabolite
`that displays reversible binding with an inhibitory activity
`toward BTK approximately 15 times lower than that of
`ibrutinib (Parmar et al. 2014).
`Ibrutinib has a mean peak plasma concentration
`observed at 1–2 h after administration. The mean termi-
`nal half-life is 4–13 h, with minimum drug accumulation
`after repeated dosing (<twofold) (Advani et al. 2013; Byrd
`et al. 2013; ImbruvicaTM., 2014). Population pharmacoki-
`netic (PK) analysis indicated that ibrutinib clearance is
`independent of age (Marostica et al. 2015).
`Multiple medications are administered in conjunction
`with ibrutinib for concurrent diseases including those for
`opportunistic infections. Given the prominent role of
`CYP3A in ibrutinib metabolism, drug–drug interactions
`(DDIs) that affect ibrutinib exposure and its metabolites,
`may occur when coadministered with potent CYP3A
`inhibitors or inducers. It is thus essential to understand
`the PK profile of ibrutinib when interacting with the CYP
`enzyme system, which can affect drug metabolism and
`clearance as well as, alter their safety and efficacy profile
`and/or of their active metabolites.
`This paper discusses results from three phase 1 studies
`which were undertaken to obtain a comprehensive under-
`standing of DDI between ibrutinib and CYP3A perpetra-
`tors ketoconazole (strong inhibitor) (Study 1), rifampin
`(strong inducer) (Study 2),and single-strength GFJ (classi-
`fied as a moderate inhibitor,
`specific for
`intestinal
`CYP3A) (Study 3) and their effect on ibrutinib exposure.
`Because it became clear during clinical development that
`food by itself significantly increases the relative bioavail-
`ability, Study 3 was performed in nonfasted condition. In
`this way, the CYP3A DDI could be assessed under more
`relevant conditions, and data from both fasted and non-
`fasted condition could be used in building a robust
`physiologically based pharmacokinetic (PBPK) model.
`
`Materials and methods
`
`Study population
`
`aged 18–55 years
`(nonsmokers)
`Healthy participants
`(inclusive) with body mass index 18–30 kg/m2 and body
`
`weight ≥50 kg were enrolled in all three studies. Study 1
`enrolled only men, whereas studies 2 and 3 enrolled both
`men and women.
`Participants with evidence of any clinically significant
`medical illness that could interfere with interpretation of
`study results or other abnormalities in physical examina-
`tion, clinical
`laboratory parameters, vital signs, or ECG
`abnormalities etc. detected at screening, were excluded
`from all three studies. Women were required to be post-
`menopausal or surgically sterile. In all three studies, par-
`ticipants were to refrain from taking any over-the-counter
`or prescribed medications except acetaminophen (<3 g
`per day).
`Protocols for each study were approved by an Indepen-
`dent Ethics Committee or Institutional Review Board at
`each study site and the studies were conducted in accor-
`dance with the ethical principles originating in the Decla-
`ration of Helsinki and in accordance with the ICH Good
`Clinical Practice guidelines, applicable regulatory require-
`ments, and in compliance with the protocol. All partici-
`pants provided written informed consent to participate in
`the studies.
`
`Study design and treatment
`
`Studies 1 and 2 were sequential design dedicated DDI
`studies of
`ibrutinib with ketoconazole and rifampin,
`respectively, versus Study 3, which was a two-way cross-
`over DDI study with GFJ, combined with a formal abso-
`lute bioavailability study following single oral dose
`administration of ibrutinib in comparison with a single
`intravenous (i.v.) administration. All three studies were
`single center, and open-label.
`Study 1 (clinicaltrials.gov identifier: NCT01626651)
`consisted of
`three phases: screening period (21 days),
`open-label treatment period (10 days), and follow-up per-
`(10  2 days).
`iod
`Participants
`received
`ibrutinib
`(120 mg, oral) on day 1 followed by blood sampling for
`PK analysis upto 72 h. Ketoconazole (Tara Pharmaceuti-
`cals), 400 mg oral, was given once-daily (q.d.) from days
`4 to 9 (except on day 7); on day 7 they received ibrutinib
`(40 mg, oral) in combination with ketoconazole (400 mg,
`oral) 1 h before ibrutinib dosing.
`Study 2 (clinicaltrials.gov identifier: NCT01763021)
`consisted of screening period (21 days), open-label treat-
`ment
`period
`(14 days)
`and
`follow-up
`period
`(10  2 days). Participants received ibrutinib, 560 mg,
`oral, q.d. on day 1. Rifampin (VersaPharm) 600 mg
`oral, q.d. was given from days 4 to 13 (after the last ibr-
`utinib sample collection for PK [over 72 h]). On day
`11, a second single oral dose of ibrutinib 560 mg was
`administered,
`followed by the blood sample collection
`over 72 h.
`
`2015 | Vol. 3 | Iss. 4 | e00156
`Page 2
`
`ª 2015 Janssen Research and Development, LLC. Pharmacology Research & Perspectives published by John Wiley & Sons Ltd,
`British Pharmacological Society and American Society for Pharmacology and Experimental Therapeutics.
`
`SANDOZ INC.
`
`IPR2023-00478
`
`Ex. 1036, p. 2 of 11
`
`

`

`Jan de Jong et al.
`
`CYP3A Perpetrators and Ibrutinib Exposure
`
`ibrutinib was administered after
`In studies 1 and 2,
`overnight fast; food was withheld for 4 h after ibrutinib
`administration on dosing days.
`Study 3 (clinicaltrials.gov identifier: NCT01866033) con-
`sisted of screening period (21 days), open-label treatment
`period (treatment A, B and C) (19 days), and follow-up
`period (10  2 days). All participants received ibrutinib
`(560 mg) in treatment A and then randomized to either
`treatment B (ibrutinib [560 mg] administered 30 min after
`240 mL of glucose in water) or treatment C (240 mL of
`GFJ [Albert Heijn pink] the evening before and 30 min
`before ibrutinib [140 mg]). In treatments B and C, partici-
`pants had standard breakfast 30 min after ibrutinib dosing,
`as opposed to treatment A which was given in fasted condi-
`tion. A single i.v. dose of 100 lg 13C6 PCI–32765 was
`administered 2 h after each ibrutinib oral dose.
`In all three studies, participants took ibrutinib with
`240 mL of water and lunch and subsequent standard
`meals were provided 4 h after oral ibrutinib dosing. Par-
`ticipants
`remained seated throughout morning (from
`30 min before dosing until after lunch),
`in order to
`minimize inter and intrasubject intestinal blood flow dif-
`ferences.
`
`Pharmacokinetic evaluations
`
`Sample collection
`
`Blood samples for all studies were collected by direct
`venipuncture or through an indwelling peripheral venous
`heparin lock catheter into heparin collection tubes. Sam-
`ples were centrifuged at approximately 4°C (15 min at
`1300 g); plasma was stored at ≤70°C.
`Blood samples for PK analysis were collected predose
`and 0.5, 1, 1.5, 2, 3, 4, 6, 8, 12, 16, 24, 48, and 72 h post-
`dose on days 1 and 7 (Study 1 and 3) and days 1 and 11
`(Study 2) for quantification of ibrutinib and PCI-45227.
`Additional blood samples were collected 2 h after ketoco-
`nazole dosing on day 7 (1 h after ibrutinib dosing) (Study
`1) and 2 h after rifampin dosing on day 11 (Study 2) for
`ketoconazole and rifampin measurement, respectively. In
`Study 2, samples were collected and analyzed for determi-
`nation of 4-b-hydroxycholesterol concentration on day -1,
`12 h after ibrutinib administration on day 11 and 14.
`
`Quantification range was 0.100–25.0 ng/mL for ibrutinib
`and PCI-45227 (Study 1 and 2) and 0.5–100 ng/mL (Study
`3) (de Vries et al. 2015). Quantification range for 13C6 PCI-
`32765 was 2–1000 pg/mL (de Vries et al. 2015).
`
`Pharmacokinetic analysis
`
`PK analyses were performed by noncompartmental meth-

`software Version 5.2.1
`ods using validated WinNonlin
`(Certara USA, Inc. Princeton, NJ) and Phoenix WinNonlin
`6.3. Key PK parameters included maximum observed
`plasma concentration (Cmax), time to reach maximum
`observed plasma concentration (tmax), elimination half-life
`associated with terminal slope (kz) of the semilogarithmic
`drug concentration-time curve (t1/2k), area under the
`plasma concentration-time curve (AUC) from time 0 to
`24 h (AUC24), AUC from time 0 to time of the last quanti-
`fiable concentration (AUClast), AUC from time 0 to infin-
`ity (AUC∞) and metabolite/parent ratios. In Study 1,
`apparent total clearance of drug after extravascular admin-
`istration (CL/F), and apparent volume of distribution
`based on the terminal phase (Vdz) were also estimated.
`Additionally, in Study 3, absolute bioavailability (F) and
`total clearance of drug after i.v. administration (CL) were
`also estimated.
`
`Safety evaluations
`
`treatment-
`included assessments of
`Safety evaluations
`related adverse events (AE), vital signs, 12-lead electrocar-
`diograms, clinical laboratory tests, and physical examina-
`tions. The AE severity was graded according to the
`National Cancer Institute - Common Terminology Crite-
`ria for Adverse Events (NCI-CTCAE) grading system ver-
`sion 4.03.
`
`Analysis sets
`
`Participants who had estimations of PK parameters of
`ibrutinib for both periods (ibrutinib administered alone
`and in combination with CYP3A perpetrators) were
`included in PK analysis set
`for statistical comparison.
`Participants who received at least one dose of study medi-
`cation were included in safety analysis set.
`
`Analytical methods
`Plasma concentrations of ibrutinib, its metabolite, PCI–
`45227, ketoconazole, rifampin, and 13C6 ibrutinib, were
`determined using validated analytical liquid chromatogra-
`phy-tandem mass
`spectroscopy (LC-MS/MS) methods.
`Bioanalyses were conducted at Department of Bioanalysis,
`Janssen R&D and at Frontage Laboratories, Inc., Exton, PA.
`
`Sample size determination
`
`For all three studies, sample size determinations were
`based on statistical estimation. A sample size is consid-
`ered adequate if the point estimates of GMR for PK
`parameters of primary interest (Cmax, AUCs) fall within
`the no-effect boundaries of clinical equivalence (90% CI)
`to the compound.
`
`ª 2015 Janssen Research and Development, LLC. Pharmacology Research & Perspectives published by John Wiley & Sons Ltd,
`British Pharmacological Society and American Society for Pharmacology and Experimental Therapeutics.
`
`2015 | Vol. 3 | Iss. 4 | e00156
`Page 3
`
`SANDOZ INC.
`
`IPR2023-00478
`
`Ex. 1036, p. 3 of 11
`
`

`

`CYP3A Perpetrators and Ibrutinib Exposure
`
`Jan de Jong et al.
`
`Statistical analyses
`
`All individual and mean plasma concentrations and esti-
`mated PK parameters were presented by graphic and
`descriptive statistics methods for each treatment. Linear
`mixed-effect models were applied to evaluate potential
`DDI effect. Log-transformation was performed on PK
`parameters (Cmax, AUCs) prior to the analysis, and 90%
`confidence intervals for GMR (with/without coadminis-
`tration of interacting drugs) were constructed on original
`scale.
`
`Results
`
`Subject disposition and demographics
`All enrolled participants (Study 1: n = 18; Study 3: n = 8)
`completed studies 1 and 3. In Study 2, 17/18 enrolled
`participants completed the study. One participant was
`excluded from the PK-evaluable population due to a pro-
`tocol deviation (use of prohibited concomitant pain med-
`ication). Demographics and baseline characteristics of the
`participants in three studies were consistent with the
`inclusion and exclusion criteria (Table 1). All participants
`received scheduled doses of the study drugs.
`
`Pharmacokinetic results
`
`Study 1: effect of ketoconazole on pharmacokinetics of
`ibrutinib and its metabolite
`
`Following coadministration of ibrutinib with ketoconazole
`under fasted condition, mean DN_Cmax (dose normalized
`to 120 mg) of ibrutinib increased from 11.8 to 325 ng/mL
`and mean DN_AUClast increased from 71.4 to 1599 ng h/
`
`mL (Fig. 1A). Although dose proportionality was not for-
`mally tested for ibrutinib, no deviations from linearity
`were observed neither in the Phase I escalating dose study
`nor population PK study (imbruvicaTM 2014; Marostica
`et al. 2015), thus justifying the dose-normalization of ibr-
`utinib exposure in this study. Intersubject variability in
`ibrutinib+ketoconazole treated participants for both Cmax
`and AUClast were >50% following ibrutinib administra-
`tion alone and approximately 40% when coadministered
`with ketoconazole. The Vd/F and CL/F were both lower
`following ibrutinib + ketoconazole compared with ibruti-
`nib alone (Vd/F: 885 L vs. 19049 L; CL/F: 92.0 L/h vs.
`2014 L/h, whereas there was no change in mean tmax
`(2.00 h vs. 1.75 h) and t1/2 (6.32 h vs. 8.20 h) (Table S1).
`On the other hand, dose-normalized PCI-45227 expo-
`sure was lower following coadministration with ketoconaz-
`ole
`compared with ibrutinib
`administration alone
`(Fig. 1B). The DN_Cmax was 2.6 times lower (11.1 ng/mL
`
`Table 1. Demographics and baseline characteristics (safety analysis
`set).
`
`Sex, n
`Women
`Men
`Race, n (%)
`White
`Black or African American
`Other/multiple
`Ethnicity, n (%)
`Not Hispanic or Latino
`Hispanic or Latino
`Age (years)
`Mean (SD)
`Baseline weight (kg)
`Mean (SD)
`Baseline BMI (kg/m2)
`Mean (SD)
`
`Study 11
`(n = 18)
`
`Study 22
`(n = 18)
`
`Study 33
`(n = 8)
`
`–
`
`18
`
`4 (22)
`11 (61)
`2 (11)
`
`16 (89)
`2 (11)
`
`8
`10
`
`5 (28)
`11 (61)
`2 (11)
`
`16 (89)
`2 (11)
`
`5
`3
`
`8 (100)
`
`8 (100)
`0
`
`–
`–
`
`33.7 (8.8)
`
`41.1 (11.6)
`
`46.4 (8.1)
`
`78.0 (7.4)
`
`77.6 (8.7)
`
`70.0 (13.2)
`
`26.1 (2.3)
`
`26.4 (2.5)
`
`23.5 (2.7)
`
`BMI, body mass index; GFJ, grapefruit juice; SD, standard deviation.
`1Study 1: ibrutinib + ketoconazole.
`2Study 2: ibrutinib + rifampin.
`3Study 3: Ibrutinib + grapefruit juice.
`
`vs. 29.1 ng/mL) and DN_AUClast was 1.2 times lower
`(256 ng h/mL vs. 304 ng h/mL) following ibrutinib + ke-
`toconazole treatment compared with ibrutinib alone. In-
`tersubject variability for both Cmax and AUClast was
`approximately 40% following ibrutinib administration
`alone and approximately 20% following ibrutinib + ke-
`toconazole. Tmax and t1/2 were both slightly longer follow-
`ing ibrutinib + ketoconazole (Tmax: 4.00 h vs. 2.00 h; t1/2
`18.00 h vs. 11.41 h). Following coadministration with ke-
`toconazole, ibrutinib mean DN_Cmax increased approxi-
`mately 29-fold (geometric mean [GM]: 285.49 vs. 10.0 ng/
`mL), and mean DN_AUClast increased approximately 24-
`fold (GM: 1463.43 vs. 61.16 ng h/mL) (Table 2). Mean
`metabolite to parent ratios decreased from 2.64 to 0.05 for
`Cmax and from 5.03 to 0.19 for AUC24 following ibruti-
`nib+ketoconazole coadministration.
`Plasma concentrations for ketoconazole at 1 h after
`drug intake on day 7 ranged from 231 to 15,800 ng/mL.
`Two participants had very low ketoconazole concentra-
`tions (352 ng/mL and 231 ng/mL); the interaction was
`still within the observed range.
`
`Study 2: effect of rifampin on the pharmacokinetics of
`ibrutinib and its metabolite
`
`Following coadministration with rifampin under fasted
`ibrutinib mean Cmax and AUClast decreased
`condition,
`(Cmax: 42.1 ng/mL to 3.38 ng/mL; AUClast: 335 ng h/mL
`
`2015 | Vol. 3 | Iss. 4 | e00156
`Page 4
`
`ª 2015 Janssen Research and Development, LLC. Pharmacology Research & Perspectives published by John Wiley & Sons Ltd,
`British Pharmacological Society and American Society for Pharmacology and Experimental Therapeutics.
`
`SANDOZ INC.
`
`IPR2023-00478
`
`Ex. 1036, p. 4 of 11
`
`

`

`Jan de Jong et al.
`
`CYP3A Perpetrators and Ibrutinib Exposure
`
`(A)
`
`(B)
`
`Figure 1. Dose-normalized mean (SD) logarithmic-linear plasma concentration-time profiles following oral administration of ibrutinib (120 mg)
`alone (day 1) and in combination with ketoconazole (40 mg Ibrutinib+400 mg Ketoconazole) (day 7) to healthy men. (A) Ibrutinib; (B) PCI-45227.
`374 ng h/mL (Fig. 2B). Intersubject variability for both
`to 38.0 ng h/mL) compared with ibrutinib administration
`Cmax and AUClast was greater than 30% following ibruti-
`alone (Fig. 2A). Intersubject variability for both Cmax and
`AUClast was greater than 60% following ibrutinib admin-
`nib administration alone and greater than 20% following
`ibrutinib + rifampin. Similar to the parent, median tmax
`istration alone and greater than 70% following ibruti-
`of the metabolite was delayed following ibrutinib + rifam-
`nib+rifampin. Median tmax was delayed from 1.76 to
`3.00 h. Terminal t1/2 was similar; due to multiple data
`pin coadministration compared with ibrutinib treatment
`alone (from 2.02 to 3.00 h). Terminal t1/2 trended shorter
`points below the quantification limit in the elimination
`for the combination treatment. The GMR for Cmax and
`phase, it could only be calculated for 5 of 17 participants.
`AUClast was 7.94% and 10.44% (or a 13- and 10-fold
`Following coadministration with rifampin, PCI-45227
`decrease), respectively, for ibrutinib + rifampin compared
`mean Cmax decreased from 70.0 ng/mL to 49.9 ng/mL
`and mean AUClast decreased from 946 ng h/mL to
`with ibrutinib alone (Table 2). Metabolite to parent ratio
`
`ª 2015 Janssen Research and Development, LLC. Pharmacology Research & Perspectives published by John Wiley & Sons Ltd,
`British Pharmacological Society and American Society for Pharmacology and Experimental Therapeutics.
`
`2015 | Vol. 3 | Iss. 4 | e00156
`Page 5
`
`SANDOZ INC.
`
`IPR2023-00478
`
`Ex. 1036, p. 5 of 11
`
`

`

`CYP3A Perpetrators and Ibrutinib Exposure
`
`Jan de Jong et al.
`
`Table 2. Geometric mean ratio and the 90% CI of the combination treatment (studies 1, 2, and 3) over ibrutinib.
`
`Parameter
`
`Ibrutinib + Ketoconazole4
`Cmax (ng/mL)1,2
`AUC24 (ng h/mL)1,2
`AUClast (ng h/mL)1,2
`AUC∞ (ng h/mL)1,2
`
`Ibrutinib + Rifampin5
`Cmax (ng/mL)2
`AUC24h (ng h/mL)2
`
`AUClast (ng.h/mL)2
`AUC∞ (ng h/mL)2
`
`Ibrutinib + Grapefruit Juice6
`Cmax (ng/mL)2,3
`AUC24 (ng h/mL)2,3
`AUClast (ng h/mL)2,3
`AUC∞ (ng h/mL)2,3
`
`Test treatment/reference
`treatment7
`
`Ibrutinib + Ketoconazole
`Ibrutinib
`Ibrutinib + Ketoconazole
`Ibrutinib
`Ibrutinib + Ketoconazole
`Ibrutinib
`Ibrutinib + Ketoconazole
`Ibrutinib
`
`Ibrutinib + Rifampin
`Ibrutinib
`Ibrutinib + Rifampin
`Ibrutinib
`Ibrutinib + Rifampin
`Ibrutinib
`Ibrutinib + Rifampin
`Ibrutinib
`
`Ibrutinib + GFJ
`Ibrutinib
`Ibrutinib + GFJ
`Ibrutinib
`Ibrutinib + GFJ
`Ibrutinib
`Ibrutinib + GFJ
`Ibrutinib
`
`N
`
`18
`18
`18
`18
`18
`18
`12
`12
`
`17
`17
`17
`17
`17
`17
`4
`4
`
`8
`8
`7
`7
`8
`8
`7
`7
`
`Geometric
`mean
`
`Ratio: (%)8
`
`90% CI (%)8
`
`Intrasubject
`CV (%)
`
`286
`10
`1390
`56
`1463
`61
`1860
`71
`
`3
`32
`23
`214
`28
`267
`46
`300
`
`437
`121
`1337
`529
`1236
`588
`1378
`643
`
`2854.5
`
`2480.1
`
`2392.8
`
`2620.2
`
`7.9
`
`10.9
`
`10.4
`
`15.2
`
`360.4
`
`252.9
`
`210.2
`
`214.5
`
`2397–3400
`
`2002–3073
`
`1901–3012
`
`1996–3440
`
`6–12
`
`8–15
`
`7–15
`
`5–46
`
`269–483
`
`219–293
`
`182–243
`
`184–250
`
`31
`
`38
`
`41
`
`39
`
`69
`
`61
`
`62
`
`74
`
`31
`
`13
`
`15
`
`14
`
`CV, coefficient of variation; GFJ, grapefruit juice.
`1Parameter values were natural log (ln) transformed and dose normalized to 120 mg ibrutinib before analysis.
`2A mixed-effect model with treatment as a fixed effect and participant as a random effect was used. Parameter values were natural log (ln) trans-
`formed before analysis.
`3The oral ibrutinib with grapefruit juice treatment group was dose normalized to 560 mg.
`4Ibrutinib: 40 mg and ketoconazole: 400 mg.
`5Ibrutinib: 560 mg and rifampin: 600 mg.
`6Ibrutinib: 560 mg and GFJ: 240 mL.
`7Test Treatment: ibrutinib + ketoconazole/ibrutinib + rifampin/ibrutinib + grapefruit juice, Reference Treatment: ibrutinib.
`8Ratio of parameter means (expressed as a percent) and 90% CIs were transformed back to the linear scale.
`
`for Cmax increased from 2.09 to 20.80, and the ratio for
`AUClast increased from 3.10 to 15.50.
`Plasma concentrations for rifampin on day 11, 2 h after
`drug intake ranged from 289 ng/mL to 18400 ng/mL
`(mean  SD = 9247  4957 ng/mL). Low concentration
`of 289 ng/mL did not adversely affect
`induction, as
`decrease
`in ibrutinib exposure on day 11 in this
`participant was comparable with that observed in other
`participants. Compared with predose
`values
`(39.9
`[15.50]) ng/mL), 4-b-hydroxycholesterol
`concentrations
`increased following multiple once-daily oral administra-
`tions of
`rifampin, providing evidence that
`sufficient
`induction of CYP3A had occurred when 560 mg ibrutinib
`(day 11) was administered after 1 week of
`rifampin
`600 mg q.d., and that induction was maintained until the
`
`72-h PK sample was collected on day 14 (day 11: 97.3
`[29.4] ng/mL and day 14: 119 [39.6] ng/mL).
`
`Study 3: effect of GFJ on ibrutinib exposure
`
`Pretreatment with GFJ increased ibrutinib concentrations
`in plasma. DN_Cmax of ibrutinib increased by 3.5-fold,
`whereas DN_AUC increased by 2.2-fold in presence of
`single-strength GFJ compared with oral administration of
`560 mg ibrutinib without GFJ under nonfasted condition
`(Fig. 2C). On the other hand, the AUCs of ibrutinib fol-
`lowing i.v. administration under nonfasted conditions
`with and without GFJ were the same. Mean Cmax was
`slightly higher with GFJ, but intrasubject variability was
`very high (84%) (Fig. 3).
`
`2015 | Vol. 3 | Iss. 4 | e00156
`Page 6
`
`ª 2015 Janssen Research and Development, LLC. Pharmacology Research & Perspectives published by John Wiley & Sons Ltd,
`British Pharmacological Society and American Society for Pharmacology and Experimental Therapeutics.
`
`SANDOZ INC.
`
`IPR2023-00478
`
`Ex. 1036, p. 6 of 11
`
`

`

`Jan de Jong et al.
`
`(A)
`
`CYP3A Perpetrators and Ibrutinib Exposure
`
`(B)
`
`(C)
`
`(D)
`
`Figure 2. Mean (SD)
`logarithmic-linear plasma concentration-time profiles of ibrutinib (560 mg) and metabolite PCI-45227 in absence and
`presence of CYP perpetrators: Rifampin (600 mg) (A and B: fasted condition) Grapefruit Juice (C and D: nonfasted condition).
`
`without GFJ were comparable. Cmax metabolite-to-parent
`ratio decreased from 1.03 to 0.32 with the addition of GFJ.
`When ibrutinib was orally administered with GFJ,
`apparent clearance, CL/F and half-life both decreased by
`half; CL after i.v. administration, however, was unchanged
`with or without GFJ. The CL following i.v. administration
`under nonfasted condition was higher than that under
`fasted condition.
`The magnitude of observed DDI was higher with
`higher baseline clearance. The higher the baseline CL/F,
`the larger the effect of ketoconazole. This trend was not
`observed clearly for the weaker (and intestine-specific)
`inhibitor GFJ, or for the inducer rifampin (Fig. 4).
`
`Safety
`
`Figure 3. Mean (SD) AUClast and Cmax of ibrutinib following oral
`administration
`of
`ibrutinib
`alone
`and
`in
`combination with
`ketoconazole, rifampin, or grapefruit juice; data dose normalized to
`560 mg.
`
`There was no effect of GFJ on tmax of PCI-45227
`(Fig. 2D). Dose-normalized PCI-45227 concentrations and
`AUCs following oral administration of ibrutinib with or
`
`Treatments were generally well-tolerated in all three stud-
`ies. In Study 1, only one (6%) participant reported ≥1 AE
`(musculoskeletal discomfort) after ibrutinib administra-
`tion alone, and six participants (33%) reported ≥1 AE
`
`ª 2015 Janssen Research and Development, LLC. Pharmacology Research & Perspectives published by John Wiley & Sons Ltd,
`British Pharmacological Society and American Society for Pharmacology and Experimental Therapeutics.
`
`2015 | Vol. 3 | Iss. 4 | e00156
`Page 7
`
`SANDOZ INC.
`
`IPR2023-00478
`
`Ex. 1036, p. 7 of 11
`
`

`

`CYP3A Perpetrators and Ibrutinib Exposure
`
`Jan de Jong et al.
`
`Most common AEs following treatment with ibrutinib
`alone were musculoskeletal discomfort (n = 2, 11%) and
`following ibrutinib+rifampin included toothache (n = 2,
`11%) and headache (n = 3, 17%). All AE were of grade 1
`intensity except for two participants who experienced grade
`2 AEs (headache and morbilliform skin rash) following
`rifampin treatment. In Study 3, the most common AEs
`reported by >1 subject included abdominal pain, diarrhea,
`and dizziness. All events were grade 1 in severity except for
`grade 2 abdominal pain in one participant. There were no
`serious AEs, AEs leading to discontinuation or AEs that
`did not resolve at end of the study, reported in the three
`studies. There were no relevant changes in vital signs or
`12-lead ECG,
`laboratory safety or changes in physical
`examination findings in any of the studies.
`
`Discussion
`
`Ibrutinib is extensively metabolized by CYP3A and has
`low bioavailability due to extensive first-pass metabolism.
`Low bioavailability results
`in more variable PK and
`potential variability in desired therapeutic response as
`well as undesirable adverse effects. Studies reported here
`were therefore designed to understand the DDI potential
`between ibrutinib and CYP3A perpetrators (ketoconazole,
`rifampin and GFJ) and their probable effects on ibrutinib
`bioavailability in healthy participants.
`In line with in vitro data that showed 96% of micro-
`somal clearance could be attributed to CYP3A4 (Scheers
`et al. 2015), CYP3A perpetrators had a major effect on
`ibrutinib exposure, without affecting the terminal half-life.
`There was an increase in ibrutinib concentration follow-
`ing coadministration with ketoconazole; intersubject vari-
`ability was lower compared to ibrutinib alone, which can
`be explained by close to complete bioavailability in this
`situation. Though ketoconazole concentration was within
`the observed range, a low value observed in two partici-
`pants may be due to the over-expression of CYP3A in
`these two participants. These values correlated with ibr-
`utinib Cmax and AUC that were approximately 80% and
`70% lower, respectively, than the mean values, both with
`and without ketoconazole coadministration. Thus,
`the
`interaction did not differ significantly from the mean
`value,
`suggesting that
`complete
`inhibition was
`still
`obtained. In fasted condition, F was found to be 3.9%
`(90% CI = 3.06–5.02) in Study 3 (de Vries et al. 2015),
`implying that a 24-fold increase in ibrutinib AUC∞ with
`ketoconazole (Study 1 and hence a different cohort of
`participants) would result in F of approximately 73–121%
`(24 9 3.06 to 5.02%). In line with ketoconazole study
`observations, intersubject variability of ibrutinib exposure
`increased following rifampin coadministration, due to sig-
`nificantly decreased bioavailability.
`
`Figure 4. Fold-change in AUC versus baseline apparent clearance
`following oral administration of
`ibrutinib with ketoconazole or
`rifampin (both under fasted conditions) or with grapefruit juice (with
`standard meal).
`
`Table 3. Summary of treatment-emergent adverse events (AE) seen
`in more than 10% of participants in any study (safety analysis set).
`
`Study 11
`n = 18
`n (%)
`
`Study 22
`n = 18
`n (%)
`
`Study 33
`n = 8
`n (%)
`
`6 (33)
`
`7 (39)
`
`3 (38)
`
`–
`–
`–
`–
`–
`–
`–
`–
`–
`–
`–
`
`4 (22)
`–
`
`–
`–
`–
`–
`–
`–
`
`2 (11)
`2 (11)
`–
`–
`–
`
`3 (17)
`–
`
`3 (38)
`3 (38)
`1 (13)
`1 (13)
`1 (13)
`1 (13)
`
`1 (13)
`1 (13)
`2 (25)
`1 (13)
`1 (13)
`
`Number of participants
`with treatment-emergent AE
`Diarrhea
`Abdominal pain
`Dyspepsia
`Flatulence
`Vomiting
`Nausea
`Toothache
`Musculoskeletal discomfort
`Back injury
`Epicondylitis
`Dizziness
`Headache
`Hyperventilation
`
`1Study 1: ibrutinib + ketoconazole.
`2Study 2: ibrutinib + rifampin.
`3Study 3: ibrutinib + grapefruit juice.
`
`following coadministration with ketoconazole. In Study 1,
`the most common AEs (>10%) following ibrutinib+ke-
`toconazole coadministration were headache (n = 4, 22%),
`venipuncture-related hematoma and pain (n = 1, 6%),
`(n = 2,
`abdominal discomfort
`and dyspepsia
`11%)
`(Table 3). All AEs were mild in severity (grade 1), limited
`in duration, and, resolved without medical intervention.
`In Study 2, 5 participants (28%) reported ≥1AE after
`ibrutinib administration alone,
`compared with 22%
`(n = 4) following combination therapy with rifampin.
`
`2015 | Vol. 3 | Iss. 4 | e00156
`Page 8
`
`ª 2015 Janssen Research and Development, LLC. Pharmacology Research & Perspectives published by John Wiley & Sons Ltd,
`British Pharmacological Society and American Society for Pharmacology and Experimental Therapeutics.
`
`SANDOZ INC.
`
`IPR2023-00478
`
`Ex. 1036, p. 8 of 11
`
`

`

`Jan de Jong et al.
`
`CYP3A Perpetrators and Ibrutinib Exposure
`
`There was a concomitant decrease in metabolite PCI-
`45227 concentration following coadministration of ibruti-
`nib with both ketoconazole and rifampin. Decrease in
`exposure of PCI-45227 observed with rifampin may be
`attributed to further metabolism of PCI-45227 involving
`CYP3A or induction of P-gp, of which PCI-45227 is a
`substrate. Reductions in Cmax and AUC in Study 2 corre-
`lated with an increase in 4-b-hydroxycholesterol, an
`endogenous biomarker of CYP3A activity (Bjorkhem-
`Bergman et al. 2013; Marde Arrhen et al. 2013;

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


Or .

Accessing this document will incur an additional charge of $.

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

Accept $ Charge
throbber

Still Working On It

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

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

throbber

A few More Minutes ... Still Working

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

Thank you for your continued patience.

This document could not be displayed.

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

Your account does not support viewing this document.

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

Your account does not support viewing this document.

Set your membership status to view this document.

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

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

Become a Member

One Moment Please

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

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

Your document is on its way!

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

Sealed Document

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

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


Access Government Site

We are redirecting you
to a mobile optimized page.





Document Unreadable or Corrupt

Refresh this Document
Go to the Docket

We are unable to display this document.

Refresh this Document
Go to the Docket