Antineoplastic Agent (BCR-ABL Tyrosine Kinase Inhibitor).
Pharmacology: Mechanism of Action: Asciminib is an oral and potent inhibitor of ABL/BCR::ABL1 tyrosine kinase. Asciminib inhibits the ABL1 kinase activity of the BCR::ABL1 fusion protein, by specifically targeting the ABL myristoyl pocket.
Pharmacodynamics: In vitro, asciminib inhibits the tyrosine kinase activity of ABL1 at mean IC
50 values below 3 nanomolar. In patient-derived cancer cells, asciminib specifically inhibits the proliferation of cells harboring BCR::ABL1 with IC
50 values between 1 and 25 nanomolar. In cells expressing the wild-type or the T315I mutant form of BCR::ABL1, asciminib inhibits cell growth with mean IC
50 values of 0.61 ±0.21 and 7.64 ±3.22 nanomolar, respectively.
In mouse xenograft models of CML, asciminib dose-dependently inhibited the growth of tumor harbouring either the wild-type or the T315I mutant form of BCR::ABL1, with tumor regression being observed at doses above 7.5 mg/kg or 30 mg/kg twice daily, respectively.
Cardiac electrophysiology: Asciminib (Bacrelba) treatment is associated with an exposure-related prolongation of the QT interval. The correlation between asciminib concentration and the estimated maximum mean change from baseline of the QT interval with Fridericia's correction (ΔQTCF) was evaluated in 239 patients with Ph+ CML or Ph+ acute lymphoblastic leukemia (ALL) receiving Asciminib (Bacrelba) at doses ranging from 10 to 280 mg twice daily and 80 to 200 mg once daily. The estimated mean ΔQTCF was 3.35 ms (upper bound of 90% CI: 4.43 ms) for Asciminib (Bacrelba) 40 mg twice-daily dose, 3.64 ms (upper bound of 90% CI: 4.68 ms) for the 80 mg once-daily dose and 5.37 ms (upper bound of 90% CI: 6.77 ms) for the 200 mg twice-daily dose.
Clinical Studies: Newly diagnosed Ph+ CML-CP: The clinical efficacy and safety of Asciminib (Bacrelba) in the treatment of patients with newly diagnosed Philadelphia chromosome-positive myeloid leukemia in chronic phase (Ph+ CML-CP) were demonstrated in the multi-center, randomized, active-controlled and open-label phase III study ASC4FIRST.
In this study, a total of 405 patients were randomized in a 1:1 ratio to receive either Asciminib (Bacrelba) or investigator selected tyrosine kinase inhibitors (IS-TKIs).
Prior to randomization, the investigator selected the TKI (imatinib or second generation [2G] TKI) to be used in the event of randomization to the comparator arm, based on patient characteristics and comorbidities. Patients were stratified according to EUTOS long-term survival (ELTS) risk group (low, intermediate, high), and pre-randomization selection of TKI (imatinib or 2G TKIs stratum). Patients received either Asciminib (Bacrelba) or IS-TKIs, and continued treatment until unacceptable toxicity or treatment failure occurred.
Patients were 36.8% female and 63.2% male, with median age 51 years (range: 18 to 86 years), Of the 405 patients, 23.5% were 65 years or older, while 6.2% were 75 years or older. Patients were Caucasian (53.8%), Asian (44,4%), Black (1%) and 0.7% unknown. The demographic characteristics within the imatinib (N=203) and the 2G TKIs (N=202) strata were: Median age: 55 years and 43 years, respectively; ELTS high risk group: 8.4% and 13.9%, respectively; Framingham cardiovascular disease high risk group: 35.5% and 17.8%, respectively.
The demographic characteristics were balanced across Asciminib (Bacrelba) and IS-TKIs, as well as across the two arms within the imatinib and 2G TKIs strata.
Of the 405 patients, 200 received Asciminib (Bacrelba), while 201 received IS-TKIs. Of the 201 patients receiving IS-TKIs, 99 received imatinib, 49 received nilotinib, 42 received dasatinib, and 11 received bosutinib, Four patients did not receive any treatment.
The median duration of treatment was 69.8 weeks (range: 0.7 to 107.7 weeks) for patients receiving Asciminib (Bacrelba) and 64.3 weeks (range: 1.3 to 103.1 weeks) for patients receiving IS-TKIs, By 48 weeks, 90% of patients on Asciminib (Bacrelba) and 80.6% of patients on IS-TKIs were still receiving treatment.
The study had multiple primary objectives assessing major molecular response rate (MMR) at 48 weeks. One primary objective evaluated Asciminib (Bacrelba) compared to IS-TKls. The other primary objective evaluated Asciminib (Bacrelba) compared to IS-TKIs, within the imatinib stratum. A secondary objective evaluated MMR at 48 weeks evaluating Asciminib (Bacrelba) compared to IS-TKIs, within the 2G TKIs stratum. The main efficacy outcomes from ASC4FIRST are summarized in Table 1. (See Table 1.)
Click on icon to see table/diagram/image
The predicted MMR rate at 48 weeks for the Asciminib (Bacrelba) 40 mg twice-daily dose is comparable to the MMR rate at 48 weeks observed in ASC4FIRST with the Asciminib (Bacrelba) 80 mg once-daily dose, based on exposure-response analysis.
Median time to MMR in patients receiving Asciminib (Bacrelba), IS-TKIs, IS-TKIs within the imatinib stratum, and IS-TKIs within the 2G TKIs stratum were: 24.3 weeks (95% CI: 24.1 to 24.6 weeks), 36.4 weeks (95% CI: 36.1 to 48.6 weeks), 48.6 weeks (95% CI: 36.1 to 59.6 weeks), and 36.1 weeks (95% CI: 24.4 to 48.1 weeks), respectively.
MMR rates at 48 weeks by ELTS risk group in patients receiving Asciminib (Bacrelba), IS-TKIs, IS-TKIs within the imatinib stratum, and IS-TKIs within the 2G TKIs stratum were: 72.1%, 57.6%, 50% and 65.6% for low risk, respectively; 64.3%, 36.8%, 26.7% and 48.2% for intermediate risk, respectively; 52.2%, 31.8%, 12.5% and 42.9% for high risk, respectively.
By 48 weeks, MR4.0 achieved by patients receiving Asciminib (Bacrelba), IS-TKIs, IS-TKIs within the imatinib stratum, and IS-TKIs within the 2G TKIs stratum was: 40.8%, 22.1%, 15.7%, and 28.4%, respectively. By 48 weeks, MR4.5 achieved by patients receiving Asciminib (Bacrelba), IS-TKIs, IS-TKIs within the imatinib stratum and IS-TKIs within 2G TKIs stratum was: 19.9%, 11.8%, 5.9%, and 17.7%, respectively.
Ph+ CML-CP, previously treated with two or more TKIs: The clinical efficacy and safety of Asciminib (Bacrelba) in the treatment of patients with Ph+ CML-CP previously treated with two or more tyrosine kinase inhibitors were demonstrated in the multi-center, randomized, active-controlled and open-label phase III study ASCEMBL.
In this study, a total of 233 patients were randomized in a 2:1 ratio and stratified according to major cytogenetic response (MCyR) status at baseline to receive either Asciminib (Bacrelba) 40 mg twice daily (N=157) or bosutinib 500 mg once daily (N=76). Patients continued treatment until unacceptable toxicity or treatment failure occurred.
Patients with Ph+ CML-CP previously treated with two or more TKIs were 51.5% female and 48.5% male with median age 52 years (range: 19 to 83 years). Of the 233 patients, 18.9% were 65 years or older, while 2.6% were 75 years or older. Patients were Caucasian (74.7%), Asian (14.2%) and Black (4.3%). Of the 233 patients, 80.7% and 18% had Eastern Cooperative Oncology Group (ECOG) performance status 0 or 1, respectively. Patients who had previously received 2, 3, 4, 5 or more prior lines of TKIs were 48.1%, 31.3%, 14.6% and 6%, respectively. The median duration of treatment was 156 weeks (range: 0.1 to 256.3 weeks) for patients receiving Asciminib (Bacrelba) and 30.5 weeks (range: 1 to 239.3 weeks) for patients receiving bosutinib.
The primary endpoint of the study was MMR at 24 weeks and the key secondary endpoint was MMR rate at 96 weeks. MMR is defined as BCR::ABL1 ratio S0.1% by International Scale [IS]. Secondary endpoints were complete cytogenetic response rate (CCyR) at 24 and 96 weeks, defined as no metaphases in bone marrow with a minimum of 20 metaphases examined. The main efficacy outcomes from ASCEMBL are summarized in Table 2. (See Table 2.)
Click on icon to see table/diagram/image
The predicted MMR rate at 24 weeks for the Asciminib (Bacrelba) 80 mg once-daily dose is comparable to the MMR rate at 24 weeks observed in ASCEMBL with the Asciminib (Bacrelba) 40 mg twice-daily dose, based on exposure-response analysis.
In ASCEMBL, 12.7% of patients treated with Asciminib (Bacrelba) and 13.2% of patients receiving bosutinib had one or more BCR::ABL1 mutation detected at baseline. MMR at 24 weeks was observed in 35.3% and 24.8% of patients receiving Asciminib (Bacrelba) with or without any BCR::ABL1 mutation at baseline, respectively. MMR at 24 weeks was observed in 25% and 11.1% of patients receiving bosutinib with or without any mutation at baseline, respectively. The MMR rate at 24 weeks in patients in whom the randomized treatment represented the third, fourth, fifth or more line of TKI was 29.3%, 25%, and 16.1% in patients treated with Asciminib (Bacrelba) and 20%, 13.8%, and 0% in patients receiving bosutinib, respectively.
The MMR rate at 48 weeks was 29.3% (95% CI: 22.32, 37.08) in patients receiving Asciminib (Bacrelba) and 13.2% (95% CI: 6.49, 22.87) in patients receiving bosutinib.
The Kaplan Meier estimated proportion of patients receiving Asciminib (Bacrelba) and maintaining MMR for at least 120 weeks was 97% (95% CI: 88.64, 99.2).
Ph+ CML-CP harboring the T315I mutation: The clinical efficacy and safety of Asciminib (Bacrelba) in the treatment of patients with Ph+ CML-CP harboring the T315I mutation were assessed in the first in human, multicenter, open-label phase I study X2101.
In this study, a total of 185 patients with Ph+ CML-CP without (N=115) or with (N=70) the T315I mutation received Asciminib (Bacrelba) at doses ranging from 10 to 200 mg twice daily or 80 to 200 mg once daily. Among these, 48 patients with Ph+ CML-CP harboring the T315I mutation received Asciminib (Bacrelba) at a dose of 200 mg twice daily. Patients continued treatment until unacceptable toxicity or treatment failure occurred.
Patients with Ph+ CML-CP harboring the T315I mutation who received Asciminib (Bacrelba) at a dose of 200 mg twice daily were 77.1% male and 22.9% female with median age of 56.5 years (range: 26 to 86 years). Of 48 patients, 33.3% were 65 years or older, while 8.3% were 75 years or older. The patients were Caucasian (47.9%), Asian (25%) and Black (2.1%). Seventy-five percent and 25% of patients had ECOG performance status 0 or 1, respectively. Patients who had previously received 1, 2, 3, 4 and 5 or more TKIs were 16.7%, 31.3%, 35.4%, 14.6% and 2.1%, respectively. The median duration of treatment was 181.7 weeks (range: 2 to 312 weeks).
MMR by 24 weeks was achieved in 42.2% of the evaluable patients (N=45) treated with Asciminib (Bacrelba) (95% CI: 27.7-57.8%).
MMR by 96 weeks was achieved in 48.9% of the evaluable patients (N=45) treated with Asciminib (Bacrelba).
Pharmacokinetics: Absorption: Asciminib is rapidly absorbed, with median maximum plasma levels (T
max) reached 2 to 3 hours after oral administration, independent of the dose. The geometric mean (geoCV%) of C
max at steady state is 1781 ng/mL (23%) and 793 ng/mL (49%) following administration of Asciminib (Bacrelba) at 80 mg once-daily and 40 mg twice-daily doses, respectively. The geometric mean (geoCV%) of C
max at steady state is 5642 ng/mL (40%) following administration of Asciminib (Bacrelba) at 200 mg twice-daily dose. The
geometric mean (geoCV%) of AUC
tau is 5262 ng*h/mL (48%) following administration of Asciminib (Bacrelba) at 40 mg twice-daily dose.
PBPK models predict that the asciminib absorption is approximately 100%, while bioavailability is approximately 73%.
Asciminib bioavailability may be reduced by co-administration of oral medicinal products containing hydroxypropyl-β-cyclodextrin as an excipient. Co-administration of multiple doses of itraconazole oral solution containing hydroxypropyl-β-cyclodextrin at a total of 8 g per dose with a 40 mg dose of asciminib, decreased asciminib AUC
inf by 40.2% in healthy subjects.
Food effect: Food consumption decreases asciminib bioavailability, with a high-fat meal having a higher impact on asciminib pharmacokinetics than a low-fat meal. Asciminib AUC is decreased by 62.3% with a high-fat meal and by 30% with a low-fat meal compared to the fasted state, independent of the dose (see Dosage & Administration and Interactions).
Distribution: Asciminib apparent volume of distribution at steady state is 111 L, based on population pharmacokinetic analysis. Asciminib is mainly distributed to plasma, with a mean blood-to-plasma ratio of 0.58, independent of the dose. Asciminib is 97.3% bound to human plasma proteins, independent of the dose.
Biotransformation/metabolism: Asciminib is primarily metabolized via CYP3A4-mediated oxidation (36%), UGT2B7- and UGT2B17-mediated glucuronidation (13.3% and 7.8%, respectively). PBPK models predict that asciminib biliary secretion via BCRP accounts for 31.1% of its total systemic clearance. Asciminib is the main circulating component in plasma (92. 7% of the administered dose).
Elimination: Asciminib is mainly eliminated via fecal excretion, with a minor contribution of the renal route. Eighty and 11% of the asciminib dose were recovered in the feces and in the urine of healthy subjects, respectively, following oral administration of a single 80 mg dose of [
14C]-labelled asciminib. Fecal elimination of unchanged asciminib accounts for 56. 7% of the administered dose.
The oral total clearance (CL/F) of asciminib is 6.31 L/hour, based on population pharmacokinetic analysis. The accumulation half-life (T½) of asciminib is 5.2 hours at 80 mg total daily dose.
Linearity/non-linearity: Asciminib exhibits a slight dose over-proportional increase in steady-state exposure (AUC and C
max) across the dose range of 10 to 200 mg administered once or twice daily.
The geometric mean average accumulation ratio is approximately 2-fold, independent of the dose. Steady-state conditions are achieved within 3 days at the 40 mg twice-daily dose.
In vitro evaluation of drug interaction potential: CYP450 and UGT enzymes:
In vitro, asciminib reversibly inhibits CYP3A4/5, CYP2C9 and UGT1Al at plasma concentrations reached at a total daily dose of 80 mg. In addition, asciminib reversibly inhibits CYP2C8 and CYP2C19 at plasma concentrations reached at 200 mg twice-daily dose.
Transporters: Asciminib is a substrate of BCRP and P-gp. Asciminib inhibits BCRP, P-gp, OATP1B1, OATP1B3, and OCT1 with Ki values of 24.3, 21.7, 2.46, 1.92, and 3.41 micromolar, respectively. Based on PBPK models, asciminib increases the exposure to P-gp, OATP1B and BCRP substrates (see Interactions). The clinical relevance of the interaction with OCT1 is currently unknown at Asciminib (Bacrelba) 200 mg twice-daily dose.
Multiple pathways: Asciminib is metabolized by several pathways including the CYP3A4, UGT2B7 and UGT2B17 enzymes and biliary secreted by the transporter BCRP.
Medicinal products inhibiting or inducing multiple pathways may alter Asciminib (Bacrelba) exposure.
Asciminib inhibits several pathways including CYP3A4, CYP2C9, OATP1B, P-gp and BCRP. Asciminib (Bacrelba) may increase the exposure of medicinal products, which are substrates of these pathways (see Interactions).
Special populations: Geriatric patients (65 years of age or above): Among the 556 patients receiving Asciminib (Bacrelba) in the ASC4FIRST, ASCEMBL, and X2101 studies, 130 (23.4%) were 65 years of age or older and 31 (5.6%) were 75 years of age or older.
No overall differences in the safety or efficacy of Asciminib (Bacrelba) were observed between patients of 65 years of age or above and younger patients.
Gender/Race/Body weight: Asciminib systemic exposure is not affected by gender, race, or body weight to any clinically relevant extent.
Renal impairment: A dedicated renal impairment study including 6 subjects with normal renal function (absolute glomerular filtration rate [aGFR] ≥90 mL/min) and 8 subjects with severe renal impairment not requiring dialysis (aGFR 15 to <30 mL/min) has been conducted. Asciminib AUC
inf and C
max are increased by 56% and 8%, respectively, in subjects with severe renal impairment compared to subjects with normal renal function, following oral administration of a single 40 mg dose of Asciminib (Bacrelba) (see Dosage & Administration).
Population pharmacokinetics models indicate an increase in asciminib median steady state AUC
0-24h by 11.5% in subjects with mild to moderate renal impairment, compared to subjects with normal renal function.
Hepatic impairment: A dedicated hepatic impairment study including 8 subjects each with normal hepatic function, mild hepatic impairment (Child-Pugh A score 5 to 6), moderate hepatic impairment (Child-Pugh B score 7 to 9) or severe hepatic impairment (Child-Pugh C score 10 to 15) was conducted. Asciminib AUC
inf is increased by 22%, 3% and 66% in subjects with mild, moderate, and severe hepatic impairment, respectively, compared to subjects with normal hepatic function, following oral administration of a single 40 mg dose of Asciminib (Bacrelba) (see Dosage & Administration).
Toxicology: Non-Clinical Safety Data: Asciminib was evaluated in safety pharmacology, repeated dose toxicity, genotoxicity, reproductive toxicity and phototoxicity studies.
Safety pharmacology: In safety pharmacology studies, asciminib did not have any effect on the central nervous and respiratory systems in rats at doses up to 600 mg/kg/day.
In an
in vitro study, asciminib inhibited the human ether-à-go-go-related gene (hERG) channels with an IC
50 of 11.4 micromolar. This value translates into a clinical safety margin at least 200-fold, 100-fold, or 30-fold higher when compared to asciminib free C
max in patients at the 40 mg twice-daily, 80 mg once-daily, or 200 mg twice-daily doses, respectively.
Moderate cardiovascular effects (increased heart rate, decreased systolic pressure, decreased mean arterial pressure, and decreased arterial pulse pressure) were observed in
in vivo cardiac safety studies in dogs. No QTc prolongation was evident in dogs up to the highest asciminib free exposure of 6.3 micromolar.
Repeat dose toxicity: Repeat dose toxicity studies identified the pancreas, liver, hematopoietic system, adrenal gland, and gastro-intestinal tract as target organs of asciminib.
Pancreatic effects (serum amylase and lipase increases, acinar cell lesions) occurred in dogs at AUC exposures below those achieved in patients on 40 mg twice daily, 80 mg once daily or 200 mg twice daily. A trend towards recovery was observed.
Elevations in liver enzymes and/or bilirubin were observed in rats, dogs, and monkeys. Histopathological hepatic changes (centrilobular hepatocyte hypertrophy, slight bile duct hyperplasia, increased individual hepatocyte necrosis and diffuse hepatocellular hypertrophy) were seen in rats and monkeys. These changes occurred at AUC exposures either equivalent to (rats) or 8- to 18-fold (dogs and monkeys) higher than those achieved in patients on 40 mg
twice daily or 80 mg once daily. AUC exposures were below (rats), equivalent (dogs) or approximately 2-fold higher (monkeys) than the exposure in patients on 200 mg twice daily. These changes were fully reversible.
Effects on the hematopoietic system (reduction in red blood cells mass, increased splenic or bone marrow pigment and increased reticulocytes) were consistent with a mild and regenerative, extravascular, hemolytic anemia in all species. These changes occurred at AUC exposures either equivalent to (rats) or 10- to 14-fold (dogs and monkeys) higher than those achieved in patients on 40 mg twice daily or 80 mg once daily. AUC exposures were below (rats), equivalent (dogs) or approximately 2-fold higher (monkeys) than the exposure in patients on 200 mg twice daily. These changes were fully reversible.
Minimal mucosal hypertrophy/hyperplasia (increase in thickness of the mucosa with frequent elongation of villi) was present in the duodenum of rats, at AUC exposures 30-fold or 22-fold higher than those achieved in patients on 40 mg twice daily or 80 mg once daily, respectively. AUC exposure was 4-fold higher than those achieved in patients on 200 mg twice daily. This change was fully reversible.
Minimal or slight hypertrophy of the adrenal gland and mild to moderate decreased vacuolation in the zona fasciculata occurred at AUC exposures either equivalent to (monkeys) or 19- to 13-fold (rats) higher than those achieved in patients on 40 mg twice daily or 80 mg once daily, respectively. AUC exposures were below (monkeys) or 2-fold higher (rats) than the exposure in patients on 200 mg twice daily, respectively. These changes were fully reversible.
Carcinogenicity and mutagenicity: Asciminib did not have mutagenic, clastogenic or aneugenic potential neither
in vitro nor
in vivo.
In a 2-year rat carcinogenicity study, non-neoplastic proliferative changes consisting of ovarian Sertoli cells hyperplasia were observed in female animals at doses equal to or above 30 mg/kg/day. Benign Sertoli cell tumors in the ovaries were observed in female rats at the highest dose of 66 mg/kg/day. AUC exposures to asciminib in female rats at 66 mg/kg/day were generally 8-fold or 5-fold higher than those achieved in patients at the dose of 40 mg twice daily or 80 mg once daily, respectively, and equivalent to those achieved in patients at the dose of 200 mg twice daily. No asciminib-related neoplastic or hyperplastic findings were noted in male rats at any dose level.
The clinical relevance of these findings is currently unknown.
Reproductive toxicity: For information on reproductive toxicity, see Pregnancy & Lactation.
Phototoxicity: In mice, asciminib showed dose-dependent phototoxic effects starting at 200 mg/kg/day. At the NOAEL of 60 mg/kg/day, exposure based on C
max in plasma was 15-fold, 6-fold or 2-fold higher than the exposure in patients on 40 mg twice daily, 80 mg once daily, or 200 mg twice daily, respectively.