CLINICAL PHARMACOLOGY
Mechanism Of Action
Ribociclib is an inhibitor of cyclin-dependent kinase
(CDK) 4 and 6. These kinases are activated upon binding to Dcyclins and play a
crucial role in signaling pathways which lead to cell cycle progression and
cellular proliferation. The cyclin D-CDK4/6 complex regulates cell cycle
progression through phosphorylation of the retinoblastoma protein (pRb).
In vitro, ribociclib decreased pRb phosphorylation
leading to arrest in the G1 phase of the cell cycle and reduced cell
proliferation in breast cancer cell lines. In vivo, treatment with single agent
ribociclib in a rat xenograft model with human tumor cells led to decreased
tumor volumes which correlated with inhibition of pRb phosphorylation.
Letrozole is a nonsteroidal competitive inhibitor of the
aromatase enzyme system by competitively binding to the heme of the cytochrome
P450 subunit of the enzyme, resulting in a reduction of estrogen biosynthesis
in all tissues. In postmenopausal women, estrogens are mainly derived from the
action of the aromatase enzyme, which converts adrenal androgens (primarily
androstenedione and testosterone) to estrone and estradiol. The suppression of
estrogen biosynthesis in peripheral tissues and in the cancer tissue itself can
therefore be achieved by specifically inhibiting the aromatase enzyme.
In vivo studies, using patient-derived estrogen receptor
positive breast cancer xenograft models, combination of ribociclib and
antiestrogen (e.g., letrozole) resulted in increased tumor growth inhibition
compared to each drug alone.
Pharmacodynamics
Ribociclib
Cardiac Electrophysiology
Serial, triplicate ECGs were collected following a single
dose and at steady-state to evaluate the effect of ribociclib on the QTcF
interval in patients with advanced cancer. A pharmacokinetic-pharmacodynamic
analysis included a total of 997 patients treated with ribociclib at doses
ranging from 50 to 1200 mg. The analysis suggested that ribociclib causes
concentration-dependent increases in the QTcF interval. The estimated mean
change from baseline in QTcF for KISQALI 600 mg in combination with aromatase
inhibitors was 22.0 ms (90% CI: 22.0, 23.4) at the geometric mean Cmax at
steady-state [see WARNINGS AND PRECAUTIONS].
Letrozole
In postmenopausal patients with advanced breast cancer,
daily doses of 0.1 mg to 5 mg FEMARA (letrozole) suppress plasma concentrations
of estradiol, estrone, and estrone sulfate by 75% to 95% from baseline with
maximal suppression achieved within two-three days. Suppression is
dose-related, with doses of 0.5 mg and higher giving many values of estrone and
estrone sulfate that were below the limit of detection in the assays. Estrogen
suppression was maintained throughout treatment in all patients treated at 0.5
mg or higher.
Letrozole is highly specific in inhibiting aromatase
activity. There is no impairment of adrenal steroidogenesis. No
clinically-relevant changes were found in the plasma concentrations of
cortisol, aldosterone, 11-deoxycortisol, 17hydroxy-progesterone, ACTH or in
plasma renin activity among postmenopausal patients treated with a daily dose
of FEMARA 0.1 mg to 5 mg. The ACTH stimulation test performed after 6 and 12
weeks of treatment with daily doses of 0.1, 0.25, 0.5, 1, 2.5, and 5 mg did not
indicate any attenuation of aldosterone or cortisol production. Glucocorticoid
or mineralocorticoid supplementation is, therefore, not necessary.
No changes were noted in plasma concentrations of
androgens (androstenedione and testosterone) among healthy postmenopausal women
after 0.1, 0.5, and 2.5 mg single doses of FEMARA or in plasma concentrations
of androstenedione among postmenopausal patients treated with daily doses of
0.1 mg to 5 mg. This indicates that the blockade of estrogen biosynthesis does
not lead to accumulation of androgenic precursors. Plasma levels of LH and FSH
were not affected by letrozole in patients, nor was thyroid function as
evaluated by TSH levels, T3 uptake, and T4 levels.
Pharmacokinetics
Ribociclib exhibited over-proportional increases in
exposure (peak plasma concentrations (Cmax) and area under the time
concentration curve (AUC)) across the dose range of 50 mg to 1200 mg following
both single dose and repeated doses. Following repeated 600 mg once daily
administration, steady-state was generally achieved after 8 days and ribociclib
accumulated with a geometric mean accumulation ratio of 2.51 (range: 0.972 to
6.40).
Letrozole's terminal elimination half-life is about 2
days and steady-state plasma concentration after daily 2.5 mg dosing is reached
in 2-6 weeks. Plasma concentrations at steady state are 1.5 to 2 times higher
than predicted from the concentrations measured after a single dose, indicating
a slight non-linearity in the pharmacokinetics of letrozole upon daily
administration of 2.5 mg. These steady-state levels are maintained over
extended periods, however, and continuous accumulation of letrozole does not
occur.
Absorption And Distribution
Ribociclib
The time to reach Cmax (Tmax) following ribociclib
administration was between 1 and 4 hours.
Binding of ribociclib to human plasma proteins in vitro was
approximately 70% and independent of concentration (10 to 10,000 ng/mL).
Ribociclib was equally distributed between red blood cells and plasma with a
mean in vivo blood-toplasma ratio of 1.04. The apparent volume of distribution
at steady-state (Vss/F) was 1090 L based on population PK analysis.
Food Effect
Compared to the fasted state, oral administration of a
single 600 mg dose of KISQALI film-coated tablet with a high-fat, high-calorie
meal (approximately 800 to 1000 calories with ~50% calories from fat, ~35%
calories from carbohydrates, and ~15% calories from protein) had no effect on
the rate and extent of absorption of ribociclib (Cmax GMR: 1.00; 90% CI: 0.898,
1.11; AUCinf GMR: 1.06; 90% CI: 1.01, 1.12) .
Letrozole
Letrozole is rapidly and completely absorbed from the
gastrointestinal tract and absorption is not affected by food. It is
metabolized slowly to an inactive metabolite whose glucuronide conjugate is
excreted renally, representing the major clearance pathway. About 90% of
radiolabeled letrozole is recovered in urine.
Letrozole is weakly protein bound and has a large volume
of distribution (approximately 1.9 L/kg).
Metabolism And Elimination/Excretion
Ribociclib
In vitro and in vivo studies indicated ribociclib
undergoes extensive hepatic metabolism mainly via CYP3A4 in humans. Following
oral administration of a single 600 mg dose of radio-labeled ribociclib to humans,
the primary metabolic pathways for ribociclib involved oxidation (dealkylation,
C and/or N-oxygenation, oxidation (-2H)) and combinations thereof. Phase II
conjugates of ribociclib Phase I metabolites involved N-acetylation, sulfation,
cysteine conjugation, glycosylation and glucuronidation. Ribociclib was the
major circulating drug-derived entity in plasma (44%). The major circulating
metabolites included metabolite M13 (CCI284, N-hydroxylation), M4 (LEQ803,
N-demethylation), and M1 (secondary glucuronide), each representing an
estimated 9%, 9%, and 8% of total radioactivity, and 22%, 20%, and 18% of
ribociclib exposure. Clinical activity (pharmacological and safety) of
ribociclib was due primarily to parent drug, with negligible contribution from
circulating metabolites.
Ribociclib was extensively metabolized with unchanged
drug accounting for 17% and 12% in feces and urine, respectively. Metabolite
LEQ803 was a significant metabolite in excreta and represented approximately
14% and 4% of the administered dose in feces and urine, respectively. Numerous
other metabolites were detected in both feces and urine in minor amounts
(≤ 3% of the administered dose).
The geometric mean plasma effective half-life (based on
accumulation ratio) was 32.0 hours (63% CV) and the geometric mean apparent
oral clearance (CL/F) was 25.5 L/hr (66% CV) at steady-state at 600 mg in
patients with advanced cancer. The geometric mean apparent plasma terminal
half-life (T½) of ribociclib ranged from 29.7 to 54.7 hours and geometric mean
CL/F of ribociclib ranged from 39.9 to 77.5 L/hr at 600 mg across studies in
healthy subjects.
Ribociclib is eliminated mainly via feces, with a small
contribution of the renal route. In 6 healthy male subjects, following a single
oral dose of radio-labeled ribociclib, 92% of the total administered
radioactive dose was recovered within 22 days; feces was the major route of
excretion (69%), with 23% of the dose recovered in urine.
Letrozole
Metabolism to a pharmacologically-inactive carbinol
metabolite (4,4'methanol-bisbenzonitrile) and renal excretion of the
glucuronide conjugate of this metabolite is the major pathway of letrozole
clearance. Of the radiolabel recovered in urine, at least 75% was the glucuronide
of the carbinol metabolite, about 9% was two unidentified metabolites, and 6%
was unchanged letrozole.
In human microsomes with specific CYP isozyme activity,
CYP3A4 metabolized letrozole to the carbinol metabolite while CYP2A6 formed
both this metabolite and its ketone analog. In human liver microsomes,
letrozole inhibited CYP2A6 and inhibited CYP2C19, however, the clinical
significance of these findings is unknown.
Specific Populations
Patients With Hepatic Impairment
Ribociclib
Based on a pharmacokinetic trial in patients with hepatic
impairment, mild (Child-Pugh class A) hepatic impairment had no effect on the
exposure of ribociclib. The mean exposure for ribociclib was increased less
than 2-fold in patients with moderate (Child-Pugh class B; geometric mean ratio
[GMR]: 1.44 for Cmax; 1.28 for AUCinf) and severe (Child-Pugh class C; GMR:
1.32 for Cmax; 1.29 for AUCinf) hepatic impairment. Based on a population
pharmacokinetic analysis that included 160 patients with normal hepatic
function and 47 patients with mild hepatic impairment, mild hepatic impairment
had no effect on the exposure of ribociclib, further supporting the findings
from the dedicated hepatic impairment study.
Letrozole
The effect of hepatic impairment on FEMARA exposure in
noncirrhotic cancer patients with elevated bilirubin levels has not been
determined.
In a study of subjects with mild to moderate
non-metastatic hepatic dysfunction (e.g., cirrhosis, Child-Pugh classification
A and B), the mean AUC values of the volunteers with moderate hepatic
impairment were 37% higher than in normal subjects, but still within the range
seen in subjects without impaired function.
In a pharmacokinetic study, subjects with liver cirrhosis
and severe hepatic impairment (Child-Pugh classification C, which included
bilirubins about 2-11 times ULN with minimal to severe ascites) had twofold
increase in exposure (AUC) and 47% reduction in systemic clearance. Breast
cancer patients with severe hepatic impairment are thus expected to be exposed
to higher levels of letrozole than patients with normal liver function
receiving similar doses of this drug [see CLINICAL PHARMACOLOGY].
Patients With Renal Impairment
Ribociclib
Mild (60 mL/min/1.73m² ≤ eGFR < 90 mL/min/1.73m²)
and moderate (30 mL/min/1.73m² ≤ eGFR < 60 mL/min/1.73m²) renal
impairment had no effect on the exposure of ribociclib based on a population PK
analysis.
The effect of renal impairment on the pharmacokinetics of
ribociclib was assessed in a renal impairment study in non-cancer subjects with
normal renal function (eGFR ≥ 90 mL/min/1.73 m²), severe renal impairment
(eGFR 15 to < 30 mL/min/1.73 m²), and End Stage Renal Disease (ESRD; eGFR
< 15 mL/min/1.73 m²). In subjects with severe renal impairment, AUCinf increased
by 1.96 fold, and Cmax increased by 1.51 fold compared to subjects with normal
renal function.
Letrozole
In a study of volunteers with varying renal function
(24-hour creatinine clearance: 9 to 116 mL/min), no effect of renal function on
the pharmacokinetics of single doses of 2.5 mg of FEMARA was found. In
addition, in a study of 347 patients with advanced breast cancer, about half of
whom received 2.5 mg FEMARA and half 0.5 mg FEMARA, renal impairment
(calculated creatinine clearance: 20 to 50 mL/min) did not affect steady-state
plasma letrozole concentrations.
Additional Pharmacokinetic Information On Ribociclib
The pharmacokinetics of ribociclib was investigated in
patients with advanced cancer following oral daily doses ranging from 50 mg to
1200 mg. Healthy subjects received single oral doses of 400 or 600 mg or
repeated daily oral doses (8 days) at 400 mg.
Effect Of Age, Weight, Gender, And Race
Population PK analysis showed that there are no
clinically relevant effects of age, body weight, gender, or race on the
systemic exposure of ribociclib.
Drug Interaction Studies
Drugs That Affect Ribociclib Plasma Concentrations
CYP3A Inhibitors
A drug interaction trial in healthy subjects was
conducted with ritonavir (a strong CYP3A inhibitor). Compared to ribociclib
alone, ritonavir (100 mg twice a day for 14 days) increased ribociclib Cmax and
AUCinf by 1.7-fold and 3.2-fold, respectively, following a single 400 mg
ribociclib dose. Cmax and AUC for LEQ803 (a prominent metabolite of LEE011,
accounting for less than 10% of parent exposure) decreased by 96% and 98%,
respectively. A moderate CYP3A4 inhibitor (erythromycin) is predicted to
increase ribociclib Cmax and AUC by 1.3-fold and 1.9-fold, respectively.
CYP3A Inducers
A drug interaction trial in healthy subjects was
conducted with rifampicin (a strong CYP3A4 inducer). Compared to ribociclib
alone, rifampicin (600 mg daily for 14 days) decreased ribociclib Cmax and AUCinf
by 81% and 89%, respectively, following a single 600 mg ribociclib dose. LEQ803
Cmax increased 1.7-fold and AUCinf decreased by 27%, respectively. A moderate CYP3A
inducer (efavirenz) is predicted to decrease ribociclib Cmax and AUC by 37% and
60%, respectively.
Drugs That Are Affected By KISQALI
CYP3A4 And CYP1A2 Substrates
A drug interaction trial in healthy subjects was
conducted as a cocktail study with midazolam (sensitive CYP3A4 substrate) and
caffeine (sensitive CYP1A2 substrate). Compared to midazolam and caffeine
alone, multiple doses of ribociclib (400 mg once daily for 8 days) increased
midazolam Cmax and AUCinf by 2.1fold and 3.8-fold, respectively. Administration
of ribociclib at 600 mg once daily is predicted to increase midazolam Cmax and
AUC by 2.4-fold and 5.2-fold, respectively. The effect of multiple doses of 400
mg ribociclib on caffeine was minimal, with Cmax decreased by 10% and AUCinf increased
slightly by 20%. Only weak inhibitory effects on CYP1A2 substrates are
predicted at 600 mg ribociclib once daily dose.
Gastric pH-Elevating Agents
Coadministration of ribociclib with drugs that elevate
the gastric pH was not evaluated in a clinical trial; however, altered
ribociclib absorption was not identified in a population PK analysis and was
not predicted using physiology based PK models.
Letrozole
Data from a clinical trial in patients with breast cancer
and population PK analysis indicated no drug interaction between ribociclib and
letrozole following coadministration of the drugs.
Anastrozole
Data from a clinical trial in patients with breast cancer
indicated no clinically relevant drug interaction between ribociclib and
anastrozole following coadministration of the drugs.
Exemestane
Data from a clinical trial in patients with breast cancer
indicated no clinically relevant drug interaction between ribociclib and
exemestane following coadministration of the drugs.
Fulvestrant
Data from a clinical trial in patients with breast cancer
indicated no clinically relevant effect of fulvestrant on ribociclib exposure
following coadministration of the drugs.
Tamoxifen
KISQALI is not indicated for concomitant use with
tamoxifen. Data from a clinical trial in patients with breast cancer indicated
that tamoxifen Cmax and AUC increased approximately 2-fold following
coadministration of 600 mg ribociclib.
In vitro Studies
Effect Of ribociclib On CYP Enzymes
In vitro, ribociclib was a reversible inhibitor of
CYP1A2, CYP2E1 and CYP3A4/5 and a time-dependent inhibitor of CYP3A4/5, at
clinically relevant concentrations. In vitro evaluations indicated that KISQALI
has no potential to inhibit the activities of CYP2A6, CYP2B6, CYP2C8, CYP2C9,
CYP2C19, and CYP2D6 at clinically relevant concentrations. It has no potential
for time-dependent inhibition of CYP1A2, CYP2C9, and CYP2D6, and no induction
of CYP1A2, CYP2B6, CYP2C9 and CYP3A4 at clinically relevant concentrations.
Effect Of Ribociclib On Transporters
In vitro evaluations indicated that KISQALI has a low
potential to inhibit the activities of drug transporters P-gp, OATP1B1/B3,
OCT1, MATEK2 at clinically relevant concentrations. KISQALI may inhibit BCRP,
OCT2, MATE1, and human BSEP at clinically relevant concentrations.
Effect Of Transporters On Ribociclib
Based on in vitro data, P-gp and BCRP mediated transport
are unlikely to affect the extent of oral absorption of ribociclib at
therapeutic doses. Ribociclib is not a substrate for hepatic uptake
transporters OATP1B1/1B3 or OCT-1 in vitro.
Animal Toxicology And/Or Pharmacology
Ribociclib
In vivo cardiac safety studies in dogs demonstrated dose
and concentration related QTc interval prolongation at an exposure similar to
patients receiving the recommended dose of 600 mg. There is a potential to
induce incidences of premature ventricular contractions (PVCs) at elevated
exposures (approximately 5-fold the anticipated clinical Cmax).
Clinical Studies
MONALEESA-2: KISQALI In Combination With Letrozole
Postmenopausal women with HR-positive, HER2-negative
advanced or metastatic breast cancer for initial endocrine based therapy
MONALEESA-2 (NCT01958021) was a randomized, double-blind,
placebo-controlled, multicenter clinical study of KISQALI plus letrozole versus
placebo plus letrozole conducted in postmenopausal women with HR-positive,
HER2negative, advanced breast cancer who received no prior therapy for advanced
disease.
A total of 668 patients were randomized to receive either
KISQALI and letrozole (n = 334) or placebo and letrozole (n = 334), stratified
according to the presence of liver and/or lung metastases. Letrozole 2.5 mg was
given orally once daily for 28 days, with either KISQALI 600 mg or placebo
orally once daily for 21 consecutive days followed by 7 days off until disease
progression or unacceptable toxicity. The major efficacy outcome measure for
the study was investigator-assessed progression-free survival (PFS) using
Response Evaluation Criteria in Solid Tumors (RECIST v1.1).
Patients enrolled in MONALEESA-2 had a median age of 62
years (range: 23 to 91) and 45% of patients were older than 65. The majority of
patients were White (82%), and all patients had an ECOG performance status of 0
or 1. A total of 47% of patients had received chemotherapy and 51% had received
antihormonal therapy in the neoadjuvant or adjuvant setting. Thirty-four
percent (34%) of patients had de novo metastatic disease, 21% had bone only
disease, and 59% had visceral disease.
The efficacy results from MONALEESA-2 are summarized in
Table 10 and Figure 1. The results shown are from a preplanned interim efficacy
analysis of PFS. Results were consistent across patient subgroups of prior
adjuvant or neoadjuvant chemotherapy or hormonal therapies, liver and/or lung
involvement, and bone-only metastatic disease. The PFS assessment based on a
blinded independent central radiological review was consistent with
investigator assessment. At the time of the PFS analysis, 6.5% of patients had
died, and overall survival data were immature.
Table 10: Efficacy Results – MONALEESA-2 (Investigator
Assessment, Intent-to-Treat Population)
|
KISQALI + letrozole |
Placebo + letrozole |
Progression-free survival |
N = 334 |
N = 334 |
Events (%) |
93 (27.8) |
150 (44.9) |
Median (months, 95% CI) |
NR (19.3 - NR) |
14.7 (13.0 - 16.5) |
Hazard Ratio (95% CI) |
0.556 (0.429 to 0.720) |
p-value |
< 0.0001a |
Overall Response Rate |
N = 256 |
N = 245 |
Patients with measurable disease (95% CI) |
52.7 (46.6, 58.9) |
37.1 (31.1, 43.2) |
a p-value estimated from one-sided log-rank
test
NR = not reached |
Figure 1: Kaplan-Meier Progression Free Survival
Curves – MONALEESA-2 (Intent-to-Treat Population)
MONALEESA-7: KISQALI In Combination
With An Aromatase Inhibitor
Pre/perimenopausal patients with HR-positive,
HER2-negative advanced or metastatic breast cancer for initial endocrine based
therapy
MONALEESA-7 (NCT02278120) was a randomized, double-blind,
placebo-controlled study of KISQALI plus either a non-steroidal aromatase
inhibitor (NSAI) or tamoxifen and goserelin versus placebo plus either a NSAI
or tamoxifen and goserelin conducted in pre/perimenopausal women with
HR-positive, HER2-negative, advanced breast cancer who received no prior
endocrine therapy for advanced disease.
A total of 672 patients were randomized to receive
KISQALI plus NSAI or tamoxifen plus goserelin (n=335) or placebo plus NSAI or
tamoxifen plus goserelin (n=337), stratified according to the presence of liver
and/or lung metastases, prior chemotherapy for advanced disease and endocrine
combination partner (tamoxifen and goserelin vs NSAI and goserelin). Among 248
patients who received KISQALI plus NSAI plus goserelin, 211 (85%) received
letrozole and 37 (15%) received anastrozole.
NSAI (letrozole 2.5 mg or anastrozole 1 mg) or tamoxifen
20 mg or were given orally once daily on a continuous daily schedule, goserelin
was administered as a sub-cutaneous injection on day 1 of each 28 day cycle,
with either KISQALI 600 mg or placebo orally once daily for 21 consecutive days
followed by 7 days off until disease progression or unacceptable toxicity. The major
efficacy outcome measure for the study was investigator-assessed
progression-free survival (PFS) using Response Evaluation Criteria in Solid
Tumors (RECIST) v1.1.
Patients enrolled in MONALEESA-7 had a median age of 44
years (range 25 to 58) and were primarily Caucasian (58%), Asian (29%), or
Black (3%). Nearly all patients (99%) had an ECOG performance status of 0 or 1.
Of the 672 patients, 33% had received chemotherapy in the adjuvant vs. 18% in
the neoadjuvant setting and 40% had received endocrine therapy in the adjuvant
vs 0.7% in the neoadjuvant setting prior to study entry. Forty percent (40%) of
patients had de novo metastatic disease, 24% had bone only disease, and 57% had
visceral disease. Demographics and baseline disease characteristics were
balanced and comparable between study arms, and endocrine combination partner.
The efficacy results from a pre-specified subgroup
analysis of 495 patients who had received KISQALI or placebo with NSAI plus
goserelin are summarized in Table 11 and Figure 2. Consistent results were
observed across endocrine combination partner (letrozole vs. anastrozole) and
in the stratification factor subgroups of disease site and prior chemotherapy
for advanced disease. Overall survival data were immature with 13% deaths.
Table 11: Efficacy Results – MONALEESA-7 (NSAI,
Investigator Assessment)
|
KISQALI + NSAI + goserelin |
Placebo + NSAI + goserelin |
Progression-free survival |
N = 248 |
N = 247 |
Events (n, %) |
92 (37.1%) |
132 (53.4%) |
Median (months, 95% CI) |
27.5 (19.1, NR) |
13.8 (12.6, 17.4) |
Hazard Ratio (95% CI) |
0.569 (0.436, 0.743) |
Overall Response Rate* |
N = 192 |
N = 199 |
Patients with measurable disease (95% CI) |
50.5 (43.4, 57.6) |
36.2 (29.5, 42.9) |
NR = not reached
* Based on confirmed responses |
Figure 2: Kaplan-Meier Progression Free Survival
Curves – MONALEESA-7 (NSAI, Investigator Assessment)