CLINICAL PHARMACOLOGY
Mechanism Of Action
Abemaciclib is an inhibitor of cyclin-dependent kinases 4 and 6 (CDK4 and CDK6). These kinases are activated upon binding to D-cyclins.
In estrogen receptor-positive (ER+) breast cancer cell lines, cyclin D1 and CDK4/6 promote phosphorylation of the retinoblastoma protein
(Rb), cell cycle progression, and cell proliferation. In vitro, continuous exposure to abemaciclib inhibited Rb phosphorylation and blocked
progression from G1 into S phase of the cell cycle, resulting in senescence and apoptosis. In breast cancer xenograft models, abemaciclib
dosed daily without interruption as a single agent or in combination with antiestrogens resulted in reduction of tumor size.
Pharmacodynamics
Cardiac Electrophysiology
Based on evaluation of the QTc interval in patients and in a healthy volunteer study, abemaciclib did not cause large mean increases (i.e.,
20 ms) in the QTc interval.
Pharmacokinetics
The pharmacokinetics of abemaciclib were characterized in patients with solid tumors, including metastatic breast cancer, and in healthy
subjects.
Following single and repeated twice daily dosing of 50 mg (0.3 times the approved recommended 150 mg dosage) to 200 mg of
abemaciclib, the increase in plasma exposure (AUC) and C was approximately dose proportional. Steady state was achieved within
5 days following repeated twice daily dosing, and the estimated geometric mean accumulation ratio was 2.3 (50% CV) and 3.2 (59% CV)
based on C and AUC, respectively.
Absorption
The absolute bioavailability of abemaciclib after a single oral dose of 200 mg is 45% (19% CV). The median T of abemaciclib is
8.0 hours (range: 4.1-24.0 hours).
Effect of Food
A high-fat, high-calorie meal (approximately 800 to 1000 calories with 150 calories from protein, 250 calories from carbohydrate, and 500
to 600 calories from fat) administered to healthy subjects increased the AUC of abemaciclib plus its active metabolites by 9% and increased
C by 26%.
Distribution
In vitro, abemaciclib was bound to human plasma proteins, serum albumin, and alpha-1-acid glycoprotein in a concentration independent
manner from 152 ng/mL to 5066 ng/mL. In a clinical study, the mean (standard deviation, SD) bound fraction was 96.3% (1.1) for
abemaciclib, 93.4% (1.3) for M2, 96.8% (0.8) for M18, and 97.8% (0.6) for M20. The geometric mean systemic volume of distribution is
approximately 690.3 L (49% CV).
In patients with advanced cancer, including breast cancer, concentrations of abemaciclib and its active metabolites M2 and M20 in
cerebrospinal fluid are comparable to unbound plasma concentrations.
Elimination
The geometric mean hepatic clearance (CL) of abemaciclib in patients was 26.0 L/h (51% CV), and the mean plasma elimination half-life
for abemaciclib in patients was 18.3 hours (72% CV).
Metabolism
Hepatic metabolism is the main route of clearance for abemaciclib. Abemaciclib is metabolized to several metabolites primarily by
cytochrome P450 (CYP) 3A4, with formation of N-desethylabemaciclib (M2) representing the major metabolism pathway. Additional
metabolites include hydroxyabemaciclib (M20), hydroxy-N-desethylabemaciclib (M18), and an oxidative metabolite (M1). M2, M18, and
M20 are equipotent to abemaciclib and their AUCs accounted for 25%, 13%, and 26% of the total circulating analytes in plasma,
respectively.
Excretion
After a single 150 mg oral dose of radiolabeled abemaciclib, approximately 81% of the dose was recovered in feces and approximately 3%
recovered in urine. The majority of the dose eliminated in feces was metabolites.
Specific Populations
Age, Gender, And Body Weight
Based on a population pharmacokinetic analysis in patients with cancer, age (range 24-91 years), gender (134 males and 856 females), and
body weight (range 36-175 kg) had no effect on the exposure of abemaciclib.
Patients With Renal Impairment
In a population pharmacokinetic analysis of 990 individuals, in which 381 individuals had mild renal impairment (60 mL/min ≤ CLcr
<90 mL/min) and 126 individuals had moderate renal impairment (30 mL/min ≤ CLcr <60 mL/min), mild and moderate renal impairment
had no effect on the exposure of abemaciclib [see Use In Specific Populations]. The effect of severe renal impairment (CLcr <30
mL/min) on pharmacokinetics of abemaciclib is unknown.
Patients With Hepatic Impairment
Following a single 200 mg oral dose of abemaciclib, the relative potency adjusted unbound AUC of abemaciclib plus its active
metabolites (M2, M18, M20) in plasma increased 1.2-fold in subjects with mild hepatic impairment (Child-Pugh A, n=9), 1.1-fold in
subjects with moderate hepatic impairment (Child-Pugh B, n=10), and 2.4-fold in subjects with severe hepatic impairment (Child-Pugh C,
n=6) relative to subjects with normal hepatic function (n=10) [see Use In Specific Populations]. In subjects with severe hepatic
impairment, the mean plasma elimination half-life of abemaciclib increased to 55 hours compared to 24 hours in subjects with normal
hepatic function.
Drug Interaction Studies
Effects Of Other Drugs On Abemaciclib
Strong CYP3A Inhibitors
Ketoconazole (a strong CYP3A inhibitor) is predicted to increase the AUC of abemaciclib by up to 16-fold.
Coadministration of 500 mg twice daily doses of clarithromycin (a strong CYP3A inhibitor) with a single 50 mg dose of VERZENIO (0.3
times the approved recommended 150 mg dosage) increased the relative potency adjusted unbound AUC of abemaciclib plus its active
metabolites (M2, M18, and M20) by 2.5-fold relative to abemaciclib alone in cancer patients.
Moderate CYP3A Inhibitors
Verapamil and diltiazem (moderate CYP3A inhibitors) are predicted to increase the relative potency adjusted
unbound AUC of abemaciclib plus its active metabolites (M2, M18, and M20) by approximately 1.6-fold and 2.4-fold, respectively.
Strong CYP3A Inducers: Coadministration of 600 mg daily doses of rifampin (a strong CYP3A inducer) with a single 200 mg dose of
VERZENIO decreased the relative potency adjusted unbound AUC of abemaciclib plus its active metabolites (M2, M18, and M20) by
approximately 70% in healthy subjects.
Moderate CYP3A Inducers
Efavirenz, bosentan, and modafinil (moderate CYP3A inducers) are predicted to decrease the relative potency
adjusted unbound AUC of abemaciclib plus its active metabolites (M2, M18, and M20) by 53%, 41%, and 29%, respectively.
Loperamide: Co-administration of a single 8-mg dose of loperamide with a single 400 mg dose of abemaciclib in healthy subjects increased
the relative potency adjusted unbound AUC of abemaciclib plus its active metabolites (M2 and M20) by 12%, which is not considered
clinically relevant.
Endocrine Therapies
In clinical studies in patients with breast cancer, there was no clinically relevant effect of fulvestrant, anastrozole,
letrozole, or exemestane on abemaciclib pharmacokinetics.
Effects Of Abemaciclib On Other Drugs
Loperamide
In a clinical drug interaction study in healthy subjects, coadministration of a single 8 mg dose of loperamide with a single
400 mg abemaciclib (2.7 times the approved recommended 150 mg dosage) increased loperamide AUC by 9% and C by 35%
relative to loperamide alone. These increases in loperamide exposure are not considered clinically relevant.
Metformin
In a clinical drug interaction study in healthy subjects, coadministration of a single 1000 mg dose of metformin, a clinically
relevant substrate of renal OCT2, MATE1, and MATE2-K transporters, with a single 400 mg dose of abemaciclib (2.7 times the approved
recommended 150 mg dosage) increased metformin AUC by 37% and C by 22% relative to metformin alone. Abemaciclib reduced
the renal clearance and renal secretion of metformin by 45% and 62%, respectively, relative to metformin alone, without any effect on
glomerular filtration rate (GFR) as measured by iohexol clearance and serum cystatin C.
Endocrine Therapies
In clinical studies in patients with breast cancer, there was no clinically relevant effect of abemaciclib on the
pharmacokinetics of fulvestrant, anastrozole, letrozole, or exemestane.
In Vitro Studies
Transporter Systems
Abemaciclib and its major active metabolites inhibit the renal transporters OCT2, MATE1, and MATE2-K at
concentrations achievable at the approved recommended dosage. The observed serum creatinine increase in clinical studies with
abemaciclib is likely due to inhibition of tubular secretion of creatinine via OCT2, MATE1, and MATE2-K [see ADVERSE REACTIONS].
Abemaciclib and its major metabolites at clinically relevant concentrations do not inhibit the hepatic uptake transporters OCT1, OATP1B1,
and OATP1B3 or the renal uptake transporters OAT1 and OAT3.
Abemaciclib is a substrate of P-gp and BCRP. Abemaciclib and its major active metabolites, M2 and M20, are not substrates of hepatic
uptake transporters OCT1, organic anion transporting polypeptide 1B1 (OATP1B1), or OATP1B3.
Abemaciclib inhibits P-gp and BCRP. The clinical consequences of this finding on sensitive P-gp and BCRP substrates are unknown.
CYP Metabolic Pathways
Abemaciclib and its major active metabolites, M2 and M20, do not induce CYP1A2, CYP2B6, or CYP3A at
clinically relevant concentrations. Abemaciclib and its major active metabolites, M2 and M20, down regulate mRNA of CYPs, including
CYP1A2, CYP2B6, CYP2C8, CYP2C9, CYP2D6 and CYP3A4. The mechanism of this down regulation and its clinical relevance are not
understood. However, abemaciclib is a substrate of CYP3A4, and time-dependent changes in pharmacokinetics of abemaciclib as a result of
autoinhibition of its metabolism was not observed.
P-gp and BCRP Inhibitors
In vitro, abemaciclib is a substrate of P-gp and BCRP. The effect of P-gp or BCRP inhibitors on the
pharmacokinetics of abemaciclib has not been studied.
Clinical Studies
VERZENIO In Combination With An Aromatase Inhibitor (Anastrozole Or Letrozole) (MONARCH 3)
Postmenopausal women with HR-positive, HER2-negative advanced or metastatic breast cancer with no prior systemic therapy in this
disease setting
MONARCH 3 was a randomized (2:1), double-blinded, placebo-controlled, multicenter study in postmenopausal women with HR-positive,
HER2-negative advanced or metastatic breast cancer in combination with a nonsteroidal aromatase inhibitor as initial endocrine-based
therapy, including patients not previously treated with systemic therapy for breast cancer.
Randomization was stratified by disease site (visceral, bone only, or other) and by prior (neo)adjuvant endocrine therapy (aromatase
inhibitor versus other versus no prior endocrine therapy). A total of 493 patients were randomized to receive 150 mg VERZENIO or
placebo orally twice daily, plus physician's choice of letrozole (80% of patients) or anastrozole (20% of patients). Patient median age was 63
years (range, 32-88 years) and the majority were White (58%) or Asian (30%). A total of 51% had received prior systemic therapy and 39%
of patients had received chemotherapy, 53% had visceral disease, and 22% had bone-only disease.
Efficacy results are summarized in Table 12 and Figure 1. PFS was evaluated according to RECIST version 1.1 and PFS assessment based
on a blinded independent radiologic review was consistent with the investigator assessment. Consistent results were observed across patient
stratification subgroups of disease site and prior (neo)adjuvant endocrine therapy. At the time of the PFS analysis, 19% of patients had died,
and overall survival data were immature.
Table 12: Efficacy Results in MONARCH 3 (Investigator Assessment, Intent-to-Treat Population)
|
VERZENIO plus
Anastrozole or Letrozole |
Placebo plus
Anastrozole or Letrozole |
Progression-Free Survival |
N=328 |
N=165 |
Number of patients with an event (n, %) |
138 (42.1) |
108 (65.5) |
Median (months, 95% CI) |
28.2 (23.5, NR) |
14.8 (11.2, 19.2) |
Hazard ratio (95% CI) |
0.540 (0.418, 0.698) |
p-value |
<0.0001 |
Objective Response for Patients with Measurable Disease |
N=267 |
N=132 |
Objective response ratea,b (n, %) |
148 (55.4) |
53 (40.2) |
95% CI |
49.5, 61.4 |
31.8, 48.5 |
Abbreviations: CI = confidence interval, NR = not reached.
a Complete response + partial response.
b Based upon confirmed responses. |
Figure 1: Kaplan-Meier Curves of Progression-Free Survival: VERZENIO plus Anastrozole or Letrozole versus Placebo plus Anastrozole or
Letrozole (MONARCH 3)
VERZENIO In Combination With Fulvestrant (MONARCH 2)
Patients with HR-positive, HER2-negative advanced or metastatic breast cancer with disease progression on or after prior adjuvant or
metastatic endocrine therapy
MONARCH 2 (NCT02107703) was a randomized, placebo-controlled, multicenter study in women with HR-positive, HER2-negative
metastatic breast cancer in combination with fulvestrant in patients with disease progression following endocrine therapy who had not
received chemotherapy in the metastatic setting. Randomization was stratified by disease site (visceral, bone only, or other) and by
sensitivity to prior endocrine therapy (primary or secondary resistance). Primary endocrine therapy resistance was defined as relapse while
on the first 2 years of adjuvant endocrine therapy or progressive disease within the first 6 months of first line endocrine therapy for
metastatic breast cancer. A total of 669 patients were randomized to receive VERZENIO or placebo orally twice daily plus intramuscular
injection of 500 mg fulvestrant on days 1 and 15 of cycle 1 and then on day 1 of cycle 2 and beyond (28-day cycles). Pre/perimenopausal
women were enrolled in the study and received the gonadotropin-releasing hormone agonist goserelin for at least 4 weeks prior to and for
the duration of MONARCH 2. Patients remained on continuous treatment until development of progressive disease or unmanageable
toxicity.
Patient median age was 60 years (range, 32-91 years), and 37% of patients were older than 65. The majority were White (56%), and 99% of
patients had an Eastern Cooperative Oncology Group (ECOG) performance status of 0 or 1. Twenty percent (20%) of patients had de novo
metastatic disease, 27% had bone-only disease, and 56% had visceral disease. Twenty-five percent (25%) of patients had primary endocrine
therapy resistance. Seventeen percent (17%) of patients were pre- or perimenopausal.
The efficacy results from the MONARCH 2 study are summarized in Table 13 and Figure 2. Median PFS assessment based on a blinded
independent radiologic review was consistent with the investigator assessment. Consistent results were observed across patient stratification
subgroups of disease site and endocrine therapy resistance. At the time of primary analysis of PFS, overall survival data were not mature
(20% of patients had died).
Table 13: Efficacy Results in MONARCH 2 (Investigator Assessment, Intent-to-Treat Population)
|
VERZENIO plus Fulvestrant |
Placebo plus Fulvestrant |
Progression-Free Survival |
N=446 |
N=223 |
Number of patients with an event (n, %) |
222 (49.8) |
157 (70.4) |
Median (months, 95% CI) |
16.4 (14.4, 19.3) |
9.3 (7.4, 12.7) |
Hazard ratio (95% CI) |
0.553 (0.449, 0.681) |
p-value |
p<.0001 |
Objective Response for Patients with Measurable Disease |
N=318 |
N=164 |
Objective response ratea (n, %) |
153 (48.1) |
35 (21.3) |
95% CI |
42.6, 53.6 |
15.1, 27.6 |
Abbreviation: CI = confidence interval.
a Complete response + partial response. |
Figure 2: Kaplan-Meier Curves of Progression-Free Survival: VERZENIO plus Fulvestrant
versus Placebo plus Fulvestrant (MONARCH 2)
VERZENIO Administered As A Monotherapy In Metastatic Breast Cancer (MONARCH 1)
Patients with HR-positive, HER2-negative Breast Cancer Who Received Prior Endocrine Therapy And 1-2 Chemotherapy Regimens In The Metastatic Setting
MONARCH 1 (NCT02102490) was a single-arm, open-label, multicenter study in women with measurable HR-positive, HER2-negative
metastatic breast cancer whose disease progressed during or after endocrine therapy, had received a taxane in any setting, and who received
1 or 2 prior chemotherapy regimens in the metastatic setting. A total of 132 patients received 200 mg VERZENIO orally twice daily on a
continuous schedule until development of progressive disease or unmanageable toxicity.
Patient median age was 58 years (range, 36-89 years), and the majority of patients were White (85%). Patients had an Eastern Cooperative
Oncology Group performance status of 0 (55% of patients) or 1 (45%). The median duration of metastatic disease was 27.6 months. Ninety
percent (90%) of patients had visceral metastases, and 51% of patients had 3 or more sites of metastatic disease. Fifty-one percent (51%) of
patients had had one line of chemotherapy in the metastatic setting. Sixty-nine percent (69%) of patients had received a taxane-based
regimen in the metastatic setting and 55% had received capecitabine in the metastatic setting. Table 14 provides the efficacy results from
MONARCH 1.
Table 14: Efficacy Results in MONARCH 1 (Intent-to-Treat Population)
|
VERZENIO 200 mg N=132 |
Investigator Assessed |
Independent Review |
Objective Response Ratea b, n (%) |
26 (19.7) |
23 (17.4) |
95% CI (%) |
13.3, 27.5 |
11.4, 25.0 |
Median Duration of Response |
8.6 months |
7.2 months |
95% CI (%) |
5.8, 10.2 |
5.6, NR |
Abbreviations: CI = confidence interval, NR = not reached.
a All responses were partial responses.
b Based upon confirmed responses. |