CLINICAL PHARMACOLOGY
Mechanism Of Action
Everolimus is an inhibitor of
mammalian target of rapamycin (mTOR), a serine-threonine kinase, downstream of
the PI3K/AKT pathway. The mTOR pathway is dysregulated in several human cancers
and in tuberous sclerosis complex (TSC). Everolimus binds to an intracellular
protein, FKBP-12, resulting in an inhibitory complex formation with mTOR
complex 1 (mTORC1) and thus inhibition of mTOR kinase activity. Everolimus
reduced the activity of S6 ribosomal protein kinase (S6K1) and eukaryotic
initiation factor 4E-binding protein (4E-BP1), downstream effectors of mTOR,
involved in protein synthesis. S6K1 is a substrate of mTORC1 and phosphorylates
the activation domain 1 of the estrogen receptor which results in
ligand-independent activation of the receptor. In addition, everolimus
inhibited the expression of hypoxia-inducible factor (e.g., HIF-1) and reduced
the expression of vascular endothelial growth factor (VEGF). Inhibition of mTOR
by everolimus has been shown to reduce cell proliferation, angiogenesis, and
glucose uptake in in vitro and/or in vivo studies.
Constitutive activation of the
PI3K/Akt/mTOR pathway can contribute to endocrine resistance in breast cancer. In
vitro studies show that estrogen-dependent and HER2+ breast cancer cells are
sensitive to the inhibitory effects of everolimus, and that combination
treatment with everolimus and Akt, HER2, or aromatase inhibitors enhances the
anti-tumor activity of everolimus in a synergistic manner.
Two regulators of mTORC1
signaling are the oncogene suppressors tuberin-sclerosis complexes 1 and 2 (TSC1,
TSC2). Loss or inactivation of either TSC1 or TSC2 leads to activation of
downstream signaling. In TSC, a genetic disorder, inactivating mutations in
either the TSC1 or the TSC2 gene lead to hamartoma formation throughout the
body as well as seizures and epileptogenesis. Overactivation of mTOR results in
neuronal dysplasia, aberrant axonogenesis and dendrite formation, increased
excitatory synaptic currents, reduced myelination, and disruption of the cortical
laminar structure causing abnormalities in neuronal development and function.
Treatment with an mTOR inhibitor in animal models of mTOR dysregulation in the
brain resulted in seizure suppression, prevention of the development of
new-onset seizures, and prevention of premature death.
Pharmacodynamics
Exposure-Response Relationship
In patients with TSC-associated
subependymal giant cell astrocytoma (SEGA), the magnitude of the reduction in
SEGA volume was correlated with the everolimus trough concentration.
In patients with TSC-associated
partial-onset seizures, the magnitude of the reduction in absolute seizure
frequency was correlated with the everolimus trough concentration.
Cardiac Electrophysiology
In a randomized,
placebo-controlled, cross-over study, 59 healthy subjects were administered a
single oral dose of AFINITOR (20 mg and 50 mg) and placebo. AFINITOR at single
doses up to 50 mg did not prolong the QT/QTc interval.
Pharmacokinetics
Absorption
After administration of AFINITOR in patients with
advanced solid tumors, peak everolimus concentrations are reached 1 to 2 hours
after administration of oral doses ranging from 5 mg to 70 mg. Following single
doses, Cmax is dose-proportional with daily dosing between 5 mg and 10 mg. With
single doses of 20 mg and higher, the increase in Cmax is less than
dose-proportional; however, AUC shows dose-proportionality over the 5 mg to 70
mg dose range. Steady-state was achieved within 2 weeks following once-daily
dosing.
In patients with TSC-associated SEGA, everolimus Cmin was
approximately dose-proportional within the dose range from 1.35 mg/m² to 14.4
mg/m².
Effect Of Food
In healthy subjects, a high-fat meal (containing
approximately 1000 calories and 55 grams of fat) reduced systemic exposure to
AFINITOR 10 mg (as measured by AUC) by 22% and the peak blood concentration
Cmax by 54%. Light-fat meals (containing approximately 500 calories and 20
grams of fat) reduced AUC by 32% and Cmax by 42%.
In healthy subjects who received 9 mg of AFINITOR
DISPERZ, high-fat meals (containing approximately 1000 calories and 55 grams of
fat) reduced everolimus AUC by 12% and Cmax by 60% and low-fat meals
(containing approximately 500 calories and 20 grams of fat) reduced everolimus AUC
by 30% and Cmax by 50%.
Relative Bioavailability
The AUCinf of everolimus was equivalent between AFINITOR
DISPERZ and AFINITOR; the Cmax of everolimus in the AFINITOR DISPERZ dosage
form was 20% to 36% lower than that of AFINITOR. The predicted trough concentrations
at steady-state were similar after daily administration.
Distribution
The blood-to-plasma ratio of everolimus, which is
concentration-dependent over the range of 5 to 5000 ng/mL, is 17% to 73%. The
amount of everolimus confined to the plasma is approximately 20% at blood
concentrations observed in cancer patients given AFINITOR 10 mg orally once
daily. Plasma protein binding is approximately 74% both in healthy subjects and
in patients with moderate hepatic impairment.
Elimination
The mean elimination half-life of everolimus is
approximately 30 hours.
Metabolism
Everolimus is a substrate of CYP3A4. Following oral
administration, everolimus is the main circulating component in human blood.
Six main metabolites of everolimus have been detected in human blood, including
three monohydroxylated metabolites, two hydrolytic ring-opened products, and a
phosphatidylcholine conjugate of everolimus. These metabolites were also
identified in animal species used in toxicity studies, and showed approximately
100-times less activity than everolimus itself.
Excretion
No specific elimination studies have been undertaken in
cancer patients. Following the administration of a 3 mg single dose of
radiolabeled everolimus in patients who were receiving cyclosporine, 80% of the
radioactivity was recovered from the feces, while 5% was excreted in the urine.
The parent substance was not detected in urine or feces.
Specific Populations
No relationship was apparent between oral clearance and
age or sex in patients with cancer.
Patients With Renal Impairment
No significant influence of creatinine clearance (25 to
178 mL/min) was detected on oral clearance (CL/F) of everolimus.
Patients With Hepatic Impairment
Compared to normal subjects, there was a 1.8-fold, 3.2-fold,
and 3.6-fold increase in AUC for subjects with mild (Child-Pugh class A),
moderate (Child-Pugh class B), and severe (Child-Pugh class C) hepatic
impairment, respectively. In another study, the average AUC of everolimus in
subjects with moderate hepatic impairment (Child-Pugh class B) was twice that
found in subjects with normal hepatic function [see DOSAGE AND
ADMINISTRATION, Use In Specific Populations].
Pediatric Patients
In patients with TSC-associated SEGA or TSC-associated
partial-onset seizures, the mean Cmin values normalized to mg/m² dose in
pediatric patients (< 18 years of age) were lower than those observed in
adults, suggesting that everolimus clearance adjusted to body surface area was
higher in pediatric patients as compared to adults.
Race Or Ethnicity
Based on a cross-study comparison, Japanese patients had
on average exposures that were higher than non-Japanese patients receiving the
same dose. Oral clearance (CL/F) is on average 20% higher in Black patients
than in White patients.
Drug Interaction Studies
Effect Of CYP3A4 And P-glycoprotein (P-gp) Inhibitors On Everolimus
Everolimus exposure increased when AFINITOR was
coadministered with:
- ketoconazole (a P-gp and strong CYP3A4 inhibitor) - Cmax
and AUC increased by 3.9- and 15-fold, respectively.
- erythromycin (a P-gp and moderate CYP3A4 inhibitor) - Cmax
and AUC increased by 2- and 4.4-fold, respectively.
- verapamil (a P-gp and moderate CYP3A4 inhibitor) - Cmax
and AUC increased by 2.3- and 3.5-fold, respectively.
Effect Of CYP3A4 And P-gp Inducers On Everolimus
The coadministration of AFINITOR with rifampin, a P-gp
and strong inducer of CYP3A4, decreased everolimus AUC by 63% and Cmax by 58%
compared to AFINITOR alone [see DOSAGE AND ADMINISTRATION].
Effect Of Everolimus On CYP3A4 Substrates
No clinically significant pharmacokinetic interactions
were observed between AFINITOR and the HMG-CoA reductase inhibitors
atorvastatin (a CYP3A4 substrate), pravastatin (a nonCYP3A4 substrate), and
simvastatin (a CYP3A4 substrate).
The coadministration of an oral dose of midazolam
(sensitive CYP3A4 substrate) with AFINITOR resulted in a 25% increase in
midazolam Cmax and a 30% increase in midazolam AUC0-inf.
The coadministration of AFINITOR with exemestane
increased exemestane Cmin by 45% and C2h by 64%; however, the corresponding
estradiol levels at steady state (4 weeks) were not different between the 2
treatment arms. No increase in adverse reactions related to exemestane was observed
in patients with hormone receptor-positive, HER2-negative advanced breast
cancer receiving the combination.
The coadministration of AFINITOR with long acting
octreotide increased octreotide Cmin by approximately 50%.
Effect Of Everolimus On Antiepileptic Drugs (AEDs)
Everolimus increased pre-dose concentrations of the
carbamazepine, clobazam, oxcarbazepine, and clobazam’s metabolite
N-desmethylclobazam by about 10%. Everolimus had no impact on pre-dose
concentrations of AEDs that are substrates of CYP3A4 (e.g., clonazepam and
zonisamide) or other AEDs, including valproic acid, topiramate, phenobarbital,
and phenytoin.
Animal Toxicology And/Or Pharmacology
In juvenile rat toxicity studies, dose-related delayed
attainment of developmental landmarks including delayed eyeopening, delayed
reproductive development in males and females and increased latency time during
the learning and memory phases were observed at doses as low as 0.15 mg/kg/day.
Clinical Studies
Hormone Receptor-Positive, HER2-Negative Breast Cancer
A randomized, double-blind, multicenter study (BOLERO-2,
NCT00863655) of AFINITOR in combination with exemestane vs. placebo in
combination with exemestane was conducted in 724 postmenopausal women with
estrogen receptor-positive, HER2-negative advanced breast cancer with
recurrence or progression following prior therapy with letrozole or
anastrozole. Randomization was stratified by documented sensitivity to prior
hormonal therapy (yes vs. no) and by the presence of visceral metastasis (yes
vs. no). Sensitivity to prior hormonal therapy was defined as either (1)
documented clinical benefit (complete response [CR], partial response [PR],
stable disease ≥ 24 weeks) to at least one prior hormonal therapy in the
advanced setting or (2) at least 24 months of adjuvant hormonal therapy prior
to recurrence. Patients were permitted to have received 0-1 prior lines of
chemotherapy for advanced disease. The major efficacy outcome measure was
progression-free survival (PFS) evaluated by RECIST (Response Evaluation
Criteria in Solid Tumors), based on investigator (local radiology) assessment.
Other outcome measures included overall survival (OS) and objective response
rate (ORR).
Patients were randomized 2:1 to AFINITOR 10 mg orally
once daily in combination with exemestane 25 mg once daily (n = 485) or to
placebo in combination with exemestane 25 mg orally once daily (n = 239). The
two treatment groups were generally balanced with respect to baseline
demographics and disease characteristics. Patients were not permitted to cross
over to AFINITOR at the time of disease progression.
The trial demonstrated a statistically significant
improvement in PFS by investigator assessment (Table 20 and Figure 1). The
results of the PFS analysis based on independent central radiological
assessment were consistent with the investigator assessment. PFS results were
also consistent across the subgroups of age, race, presence and extent of
visceral metastases, and sensitivity to prior hormonal therapy.
ORR was higher in the AFINITOR in combination with
exemestane arm vs. the placebo in combination with exemestane arm (Table 20).
There were 3 complete responses (0.6%) and 58 partial responses (12%) in the
AFINITOR arm. There were no complete responses and 4 partial responses (1.7%)
in the placebo in combination with exemestane arm.
After a median follow-up of 39.3 months, there was no
statistically significant difference in OS between the AFINITOR in combination
with exemestane arm and the placebo in combination with exemestane arm [HR 0.89
(95% CI: 0.73, 1.10)].
Table 20: Efficacy Results in Hormone-Receptor
Positive, HER-2 Negative Breast Cancer in BOLERO-2
Analysis |
AFINITOR with Exemestane
N = 485 |
Placebo with Exemestane
N = 239 |
Hazard ratio |
p-value |
Median progression-free survival (months, 95% CI) |
Investigator radiological review |
7.8
(6.9, 8.5) |
3.2
(2.8, 4.1) |
0.45a (0.38, 0.54) |
< 0.0001b |
|
Independent radiological review |
11.0
(9.7, 15.0) |
4.1
(2.9, 5.6) |
0.38a (0.3, 0.5) |
< 0.0001b |
Best overall response (%, 95% CI) |
Objective response rate (ORR)c |
12.6%
(9.8, 15.9) |
1.7%
(0.5, 4.2) |
n/ad |
|
aHazard ratio is obtained from the stratified
Cox proportional-hazards model by sensitivity to prior hormonal therapy and
presence of visceral metastasis
bp-value is obtained from the one-sided log-rank test stratified by
sensitivity to prior hormonal therapy and presence of visceral metastasis
cObjective response rate = proportion of patients with CR or PR dNot
applicable |
Figure 1: Kaplan-Meier
Curves for Progression-Free Survival by Investigator Radiological Review in
Hormone Receptor-Positive, HER-2 Negative Breast Cancer in BOLERO-2
Neuroendocrine Tumors (NET)
Pancreatic Neuroendocrine
Tumors (PNET)
A randomized, double-blind,
multi-center trial (RADIANT-3, NCT00510068) of AFINITOR in combination with
best supportive care (BSC) compared to placebo in combination with BSC was
conducted in patients with locally advanced or metastatic advanced PNET and
disease progression within the prior 12 months. Patients were stratified by
prior cytotoxic chemotherapy (yes vs. no) and WHO performance status (0
vs. 1 and 2). Treatment with somatostatin analogs was allowed as part of BSC.
The major efficacy outcome was PFS evaluated by RECIST. After documented radiological
progression, patients randomized to placebo could receive open-label AFINITOR.
Other outcome measures included ORR, response duration, and OS.
Patients were randomized 1:1 to receive either AFINITOR
10 mg once daily (n = 207) or placebo (n = 203). Demographics were well
balanced (median age 58 years, 55% male, 79% White). Of the 203 patients
randomized to BSC, 172 patients (85%) received AFINITOR following documented
radiologic progression.
The trial demonstrated a statistically significant improvement
in PFS (Table 21 and Figure 2). PFS improvement was observed across all patient
subgroups, irrespective of prior somatostatin analog use. The PFS results by
investigator radiological review, central radiological review and adjudicated
radiological review are shown below in Table 21.
Table 21: Progression-Free Survival Results in PNET in
RADIANT-3
Analysis |
AFINITOR
N = 207 |
Placebo
N = 203 |
Hazard Ratio (95% CI) |
p-value |
N 410 |
Median progression-free survival (months) (95% CI) |
Investigator radiological review |
11.0
(8.4, 13.9) |
4.6
(3.1, 5.4) |
0.35
(0.27, 0.45) |
< 0.001 |
Central radiological review |
13.7
(11.2, 18.8) |
5.7
(5.4, 8.3) |
0.38
(0.28, 0.51) |
< 0.001 |
Adjudicated radiological reviewa |
11.4
(10.8, 14.8) |
5.4
(4.3, 5.6) |
0.34
(0.26, 0.44) |
< 0.001 |
aIncludes adjudication for discrepant
assessments between investigator radiological review and central radiological
review |
Figure 2: Kaplan-Meier Curves for Progression-Free Survival by Investigator Radiological Review in
PNET in RADIANT-3
Investigator-determined
response rate was 4.8% in the AFINITOR arm and there were no complete
responses. OS was not statistically significantly different between arms [HR =
0.94 (95% CI 0.73, 1.20); p = 0.30].
NET Of Gastrointestinal (GI) Or
Lung Origin
A randomized, double-blind,
multicenter study (RADIANT-4, NCT01524783) of AFINITOR in combination with BSC
compared to placebo in combination with BSC was conducted in patients with
unresectable, locally advanced or metastatic, well differentiated,
non-functional NET of GI (excluding pancreatic) or lung origin. The study
required that patients had well-differentiated (low or intermediate grade)
histology, no prior or current history of carcinoid symptoms, and evidence of
disease progression within 6 months prior to randomization. Patients were
randomized 2:1 to receive either AFINITOR 10 mg once daily or placebo, and
stratified by prior somatostatin analog use (yes vs. no), tumor origin and WHO
performance status (0 vs. 1). The major efficacy outcome measure was PFS based
on independent radiological assessment evaluated by RECIST. Additional efficacy
outcome measures were OS and ORR.
A total of 302 patients were
randomized, 205 to the AFINITOR arm and 97 to the placebo arm. The median age
was 63 years (22 to 86 years); 47% were male; 76% were White; 74% had WHO
performance status of 0 and 26% had WHO performance status of 1. The most
common primary sites of tumor were lung (30%), ileum (24%), and rectum (13%).
The study demonstrated a
statistically significant improvement in PFS per independent radiological
review (Table 22 and Figure 3). There was no statistically significant
difference in OS at the planned interim analysis.
Table 22: Progression-Free
Survival in NET of GI or Lung Origin in RADIANT-4
|
AFINITOR
N = 205 |
Placebo
N = 97 |
Progression-Free Survival |
Number of Events |
113 (55%) |
65 (67%) |
Progressive Disease |
104 (51%) |
60 (62%) |
Death |
9 (4%) |
5 (5%) |
Median PFS in months (95% CI) |
11.0 (9.2, 13.3) |
3.9 (3.6, 7.4) |
Hazard Ratio (95% CI)a |
0.48 (0.35, 0.67) |
p-valueb |
< 0.001 |
Overall Response Rate |
2% |
1% |
a Hazard ratio is obtained from the stratified
Cox model.
b p-value is obtained from the stratified log-rank test. |
Figure 3: Kaplan-Meier Curves for Progression-Free Survival in NET of GI or Lung Origin in RADIANT-4
Lack Of Efficacy In Locally
Advanced Or Metastatic Functional Carcinoid Tumors
The safety and effectiveness of
AFINITOR in patients with locally advanced or metastatic functional carcinoid
tumors have not been demonstrated. In a randomized (1:1), double-blind,
multi-center trial (RADIANT-2, NCT00412061) in 429 patients with carcinoid
tumors, AFINITOR in combination with long-acting octreotide (Sandostatin LAR®)
was compared to placebo in combination with long-acting octreotide. After
documented radiological progression, patients on the placebo arm could receive
AFINITOR; of those randomized to placebo, 67% received open-label AFINITOR in
combination with long-acting octreotide. The study did not meet its major
efficacy outcome measure of a statistically significant improvement in PFS and
the final analysis of OS favored the placebo in combination with long-acting
octreotide arm.
Renal Cell Carcinoma (RCC)
An international, multi-center,
randomized, double-blind trial (RECORD-1, NCT00410124) comparing AFINITOR 10 mg
once daily and placebo, both in conjunction with BSC, was conducted in patients
with metastatic RCC whose disease had progressed despite prior treatment with
sunitinib, sorafenib, or both sequentially. Prior therapy with bevacizumab,
interleukin 2, or interferon-α was also permitted. Randomization was
stratified according to prognostic score and prior anticancer therapy. The
major efficacy outcome measure for the trial was PFS evaluated by RECIST, based
on a blinded, independent, central radiologic review. After documented
radiological progression, patients randomized to placebo could receive
open-label AFINITOR. Other outcome measures included OS.
In total, 416 patients were
randomized 2:1 to receive AFINITOR (n = 277) or placebo (n = 139). Demographics
were well balanced between the arms (median age 61 years; 77% male, 88% White,
74% received prior sunitinib or sorafenib, and 26% received both sequentially).
AFINITOR was superior to
placebo for PFS (Table 23 and Figure 4). The treatment effect was similar
across prognostic scores and prior sorafenib and/or sunitinib. Final OS results
yield a hazard ratio of 0.90 (95% CI: 0.71, 1.14), with no statistically
significant difference between the arms. Planned cross-over from placebo due to
disease progression to open-label AFINITOR occurred in 80% of the 139 patients
and may have confounded the OS benefit.
Table 23: Progression-Free Survival and Objective
Response Rate by Central Radiologic Review in RCC in RECORD-1
|
AFINITOR
N = 277 |
Placebo
N = 139 |
Hazard Ratio (95% CI) |
p-valuea |
Median Progression-free Survival (95% CI) |
4.9 months (4.0, 5.5) |
1.9 months (1.8, 1.9) |
0.33 (0.25, 0.43) |
< 0.0001 |
Objective Response Rate |
2% |
0% |
n/ab |
n/ab |
aLog-rank test stratified by prognostic score. |
Figure 4: Kaplan-Meier Curves for Progression-Free Survival in RCC in RECORD-1
Tuberous Sclerosis Complex
(TSC)-Associated Renal Angiomyolipoma
A randomized (2:1),
double-blind, placebo-controlled trial (EXIST-2, NCT00790400) of AFINITOR was
conducted in 118 patients with renal angiomyolipoma as a feature of TSC (n =
113) or sporadic lymphangioleiomyomatosis (n = 5). The key eligibility
requirements for this trial were at least one angiomyolipoma of ≥ 3 cm in
longest diameter on CT/MRI based on local radiology assessment, no immediate
indication for surgery, and age ≥ 18 years. Patients received AFINITOR 10
mg or matching placebo orally once daily until disease progression or
unacceptable toxicity. CT or MRI scans for disease assessment were obtained at
baseline, 12, 24, and 48 weeks and annually thereafter. Clinical and
photographic assessment of skin lesions were conducted at baseline and every 12
weeks thereafter until treatment discontinuation. The major efficacy outcome
measure was angiomyolipoma response rate based on independent central radiology
review, which was defined as a ≥ 50% reduction in angiomyolipoma volume,
absence of new angiomyolipoma lesion ≥ 1 cm, absence of kidney volume
increase ≥ 20%, and no angiomyolipoma related bleeding of ≥ Grade
2. Key supportive efficacy outcome measures were time to angiomyolipoma
progression and skin lesion response rate. The primary analyses of efficacy
outcome measures were limited to the blinded treatment period and conducted 6
months after the last patient was randomized. The comparative angiomyolipoma
response rate analysis was stratified by use of enzyme-inducing antiepileptic
drugs (EIAEDs) at randomization (yes vs. no).
Of the 118 patients enrolled,
79 were randomized to AFINITOR and 39 to placebo. The median age was 31 years
(18 to 61 years), 34% were male, and 89% were White. At baseline, 17% of
patients were receiving EIAEDs. On central radiology review at baseline, 92% of
patients had at least 1 angiomyolipoma of ≥ 3 cm in longest diameter, 29%
had angiomyolipomas ≥ 8 cm, 78% had bilateral angiomyolipomas, and
97% had skin lesions. The median values for the sum of all target renal
angiomyolipoma lesions at baseline were 85 cm³ (9 to 1612 cm³) and 120 cm³ (3
to 4520 cm³) in the AFINITOR and placebo arms, respectively. Forty-six (39%)
patients had prior renal embolization or nephrectomy. The median duration of
follow-up was 8.3 months (0.7 to 24.8 months) at the time of the primary
analysis.
The renal angiomyolipoma response rate was statistically
significantly higher in AFINITOR-treated patients (Table 24). The median
response duration was 5.3+ months (2.3+ to 19.6+ months).
There were 3 patients in the AFINITOR arm and 8 patients
in the placebo arm with documented angiomyolipoma progression by central
radiologic review (defined as a ≥ 25% increase from nadir in the sum of
angiomyolipoma target lesion volumes to a value greater than baseline,
appearance of a new angiomyolipoma ≥ 1 cm in longest diameter, an
increase in renal volume ≥ 20% from nadir for either kidney and to a
value greater than baseline, or Grade ≥ 2 angiomyolipoma-related
bleeding). The time to angiomyolipoma progression was statistically
significantly longer in the AFINITOR arm (HR 0.08 [95% CI: 0.02, 0.37]; p <
0.0001).
Table 24: Angiomyolipoma
Response Rate in TSC-Associated Renal Angiomyolipoma in EXIST-2
|
AFINITOR
N = 79 |
Placebo
N = 39 |
p-value |
Primary analysis |
Angiomyolipoma response ratea - % 95% CI |
41.8 (30.8, 53.4) |
0 (0.0, 9.0) |
< 0.0001 |
aPer independent central radiology review |
Skin lesion response rates were
assessed by local investigators for 77 patients in the AFINITOR arm and 37
patients in the placebo arm who presented with skin lesions at study entry. The
skin lesion response rate was statistically significantly higher in the
AFINITOR arm (26% vs. 0, p = 0.0011); all skin lesion responses were partial
responses, defined as visual improvement in 50% to 99% of all skin lesions
durable for at least 8 weeks (Physician's Global Assessment of Clinical
Condition).
Patients randomized to placebo
were permitted to receive AFINITOR at the time of angiomyolipoma progression or
after the time of the primary analysis. After the primary analysis, patients
treated with AFINITOR underwent additional follow-up CT or MRI scans to assess
tumor status until discontinuation of treatment or completion of 4 years of
follow-up after the last patient was randomized. A total of 112 patients (79
randomized to AFINITOR and 33 randomized to placebo) received at least one dose
of AFINITOR. The median duration of AFINITOR treatment was 3.9 years (0.5
months to 5.3 years) and the median duration of follow-up was 3.9 years (0.9
months to 5.4 years). During the follow-up period after the primary analysis,
32 patients (in addition to the 33 patients identified at the time of the
primary analysis) had an angiomyolipoma response based upon independent central
radiology review. Among the 65 responders out of 112 patients, the median time
to angiomyolipoma response was 2.9 months (2.6 to 33.8 months). Fourteen
percent of the 112 patients treated with AFINITOR had angiomyolipoma
progression by the end of the follow-up period. No patient underwent a
nephrectomy for angiomyolipoma progression and one patient underwent renal
embolization while treated with AFINITOR.
Tuberous Sclerosis Complex
(TSC)-Associated Subependymal Giant Cell Astrocytoma (SEGA)
EXIST-1
A randomized (2:1), double-blind, placebo-controlled
trial (EXIST-1, NCT00789828) of AFINITOR was conducted in 117 pediatric and
adult patients with SEGA and TSC. Eligible patients had at least one SEGA
lesion ≥ 1 cm in longest diameter on MRI based on local radiology
assessment and one or more of the following: serial radiological evidence of
SEGA growth, a new SEGA lesion ≥ 1 cm in longest diameter, or new or
worsening hydrocephalus. Patients randomized to the treatment arm received
AFINITOR at a starting dose of 4.5 mg/m² daily, with subsequent dose
adjustments as needed to achieve and maintain everolimus trough concentrations
of 5 to 15 ng/mL as tolerated. AFINITOR or matched placebo
continued until disease progression or unacceptable toxicity. MRI scans for
disease assessment were obtained at baseline, 12, 24, and 48 weeks, and
annually thereafter.
The main efficacy outcome
measure was SEGA response rate based on independent central radiology review.
SEGA response was defined as a ≥ 50% reduction in the sum of SEGA volume
relative to baseline, in the absence of unequivocal worsening of non-target SEGA
lesions, a new SEGA lesion ≥ 1 cm, and new or worsening hydrocephalus.
The primary analysis of SEGA response rate was limited to the blinded treatment
period and conducted 6 months after the last patient was randomized. The
analysis of SEGA response rate was stratified by use of enzyme-inducing
antiepileptic drugs (EIAEDs) at randomization (yes vs. no).
Of the 117 patients enrolled, 78 were randomized to
AFINITOR and 39 to placebo. The median age was 9.5 years (0.8 to 26 years); a
total of 20 patients were < 3 years, 54 patients were 3 to < 12 years, 27
patients were 12 to < 18 years, and 16 patients were ≥ 18 years; 57%
were male, and 93% were White. At baseline, 18% of patients were receiving
EIAEDs. Based on central radiology review at baseline, 98% of patients had at
least one SEGA lesion ≥ 1.0 cm in longest diameter, 79% had bilateral
SEGAs, 43% had ≥ 2 target SEGA lesions, 26% had growth in or into the
inferior surface of the ventricle, 9% had evidence of growth beyond the
subependymal tissue adjacent to the ventricle, and 7% had radiographic evidence
of hydrocephalus. The median values for the sum of all target SEGA lesions at
baseline were 1.63 cm³ (0.18 to 25.15 cm³) and 1.30 cm³ (0.32 to 9.75 cm³) in
the AFINITOR and placebo arms respectively. Eight (7%) patients had prior
SEGA-related surgery. The median duration of follow-up was 8.4 months (4.6 to
17.2 months) at the time of primary analysis.
The SEGA response rate was
statistically significantly higher in AFINITOR-treated patients (Table 25). At
the time of the primary analysis, all SEGA responses were ongoing and the
median duration of response was 5.3 months (2.1 to 8.4 months).
With a median follow-up of 8.4
months, SEGA progression was detected in 15.4% of the 39 patients randomized to
receive placebo and none of the 78 patients randomized to receive AFINITOR. No
patient in either treatment arm required surgical intervention.
Table 25: Subependymal Giant
Cell Astrocytoma Response Rate in TSC-Associated SEGA in EXIST-1
|
AFINITOR
N = 78 |
Placebo
N = 39 |
p-value |
Primary analysis |
SEGA response ratea - (%) |
35 |
0 |
< 0.0001 |
95% CI |
24, 46 |
0, 9 |
|
a Per independent central radiology review |
Patients randomized to placebo
were permitted to receive AFINITOR at the time of SEGA progression or after the
primary analysis, whichever occurred first. After the primary analysis,
patients treated with AFINITOR underwent additional follow-up MRI scans to
assess tumor status until discontinuation of treatment or completion of 4 years
of follow-up after the last patient was randomized. A total of 111 patients (78
patients randomized to AFINITOR and 33 patients randomized to placebo) received
at least one dose of AFINITOR. Median duration of AFINITOR treatment and
follow-up was 3.9 years (0.2 to 4.9 years).
By four years after the last
patient was enrolled, 58% of the 111 patients treated with AFINITOR had a
≥ 50% reduction in SEGA volume relative to baseline, including 27
patients identified at the time of the primary analysis and 37 patients with a
SEGA response after the primary analysis. The median time to SEGA response was
5.3 months (2.5 to 33.1 months). Twelve percent of the 111 patients treated
with AFINITOR had documented disease progression by the end of the follow-up
period and no patient required surgical intervention for SEGA during the study.
Study 2485
Study 2485 (NCT00411619) was an open-label, single-arm
trial conducted to evaluate the antitumor activity of AFINITOR 3 mg/m²/orally
once daily in patients with SEGA and TSC. Serial radiological evidence of SEGA
growth was required for entry. Tumor assessments were performed every 6 months
for 60 months after the last patient was enrolled or disease progression,
whichever occurred earlier. The major efficacy outcome measure was the
reduction in volume of the largest SEGA lesion with 6 months of treatment, as
assessed via independent central radiology review. Progression was defined as
an increase in volume of the largest SEGA lesion over baseline that was ≥
25% over the nadir observed on study.
A total of 28 patients received AFINITOR for a median
duration of 5.7 years (5 months to 6.9 years); 82% of the 28 patients remained
on AFINITOR for at least 5 years. The median age was 11 years (3 to 34 years),
61% male, 86% White.
At the primary analysis, 32% of the 28 patients (95% CI:
16%, 52%) had an objective response at 6 months, defined as at least a 50%
decrease in volume of the largest SEGA lesion. At the completion of the study,
the median duration of durable response was 12 months (3 months to 6.3 years).
By 60 months after the last patient was enrolled, 11% of
the 28 patients had documented disease progression. No patient developed a new
SEGA lesion while on AFINITOR. Nine additional patients were identified as
having a > 50% volumetric reduction in their largest SEGA lesion between 1
to 4 years after initiating AFINITOR including 3 patients who had surgical
resection with subsequent regrowth prior to receiving AFINITOR.
Tuberous Sclerosis Complex (TSC)-Associated Partial-Onset
Seizures
The efficacy of AFINITOR DISPERZ as an adjunctive
anti-epileptic drug (AED) was evaluated in a randomized, double-blind,
multicenter, placebo-controlled study conducted in patients with TSC-associated
partial-onset seizures (EXIST-3, NCT01713946). Patients with a history of
inadequate control of partial-onset seizures despite treatment with ≥ 2
sequential AED regimens were randomized to receive placebo or AFINITOR DISPERZ
once daily at a dose to achieve a low trough (LT) level (3-7 ng/mL) or a high
trough (HT) level (9-15 ng/mL). Randomization was stratified by age group (1 to
< 6, 6 to < 12, 12 to < 18, ≥ 18 years). The study consisted of
3 phases: an 8-week Baseline observation phase; an 18-week double-blind,
placebo-controlled Core phase (6-week titration period and a 12-week
maintenance period), and an Extension phase of ≥ 48 weeks. Patients were
required to have a diagnosis of TSC per the modified Gomez criteria, and
≥ 16 partial-onset seizures during the Baseline phase while receiving a
stable dose of 1 to 3 concomitant AEDs. The starting doses for AFINITOR DISPERZ
in the Core phase ranged from 3 to 6 mg/m² orally once daily, depending on age,
in patients not receiving concomitant CYP3A4/P-gp inducers and from 5 to 9 mg/m²
orally once daily, depending on age, in patients receiving concomitant
CYP3A4/P-gp inducers. During the 6-week titration period, everolimus trough
levels were assessed every 2 weeks and up to 3 dose adjustments were allowed to
attempt to reach the targeted everolimus trough concentration range.
The major efficacy outcome measure was the percentage
reduction in seizure frequency from the Baseline phase, during the maintenance
period of the Core phase. Additional efficacy outcome measures included
response rate, defined as at least a 50% reduction in seizure frequency from
the Baseline phase during the maintenance period of the Core phase, and seizure
freedom rate during the maintenance period of the Core phase.
A total of 366 patients were randomized to AFINITOR
DISPERZ LT (n = 117), AFINITOR DISPERZ HT (n = 130) or placebo (n = 119).
Median age was 10.1 years (2.2 to 56 years); 28% of patients were < 6 years,
31% were 6 to < 12 years, 22% were 12 to < 18 years, and 18% were ≥
18 years). The majority were White (65%) and male (52%). The most common major
features of TSC were cortical tubers (92%), hypomelanotic macules (84%), and
subependymal nodules (83%). While 17% of the patients had SEGA, 42% had renal
angiomyolipoma, and 9% had both SEGA and renal angiomyolipoma; no patients were
receiving treatment with AFINITOR or AFINITOR DISPERZ for these manifestations
of TSC. During the Baseline phase, 65% of patients had complex partial
seizures, 52% had secondarily generalized seizures, 19% had simple partial
seizures, and 2% had generalized onset seizures. The median seizure frequency
per week during the Baseline phase was 9.4 for all patients and 47% of patients
were receiving 3 AEDs during the Baseline phase. The efficacy results are
summarized in Table 26.
Table 26: Percentage Reduction in Seizure Frequency
and Response Rate in TSC-Associated Partial-Onset Seizures in EXIST-3
|
AFINITOR DISPERZ |
Placebo
N = 119 |
Target of 3-7 ng/mL
N = 117 |
Target of 9-15 ng/mL
N = 130 |
Seizures per week |
Median at Baseline (Min, Max) |
8.6
(1.4, 192.9) |
9.5
(0.3, 218.4) |
10.5
(1.3, 231.7) |
Median at Core phasea (Min, Max) |
6.8
(0.0, 193.5) |
4.9
(0.0, 133.7) |
8.5
(0.0, 217.7) |
Percentage reduction from Baseline to Core phase (Maintenance a) |
Median |
29.3 |
39.6 |
14.9 |
95% CIb |
18.8, 41.9 |
35.0, 48.7 |
0.1, 21.7 |
p-valuec |
0.003 |
< 0.001 |
|
Response rate |
Responders, n (%) |
28.2 |
40 |
15.1 |
95% CId |
20.3, 37.3 |
31.5, 49.0 |
9.2, 22.8 |
aIf patient discontinued before starting the
Maintenance period, then the Titration period is used
b95% CI of the median based on bootstrap percentiles
cp-values were for superiority vs. placebo, and obtained from rank
ANCOVA with Baseline seizure frequency as covariate, stratified by age subgroup
dExact 95% CI obtained using Clopper-Pearson method |