EMA Review of Acalabrutinib for the Treatment of Adult Patients with Chronic Lymphocytic Leukemia (2024)

  • Journal List
  • Oncologist
  • v.26(3); 2021 Mar
  • PMC7930415

As a library, NLM provides access to scientific literature. Inclusion in an NLM database does not imply endorsem*nt of, or agreement with, the contents by NLM or the National Institutes of Health.
Learn more: PMC Disclaimer | PMC Copyright Notice

EMA Review of Acalabrutinib for the Treatment of Adult Patients with Chronic Lymphocytic Leukemia (1)

Link to Publisher's site

Oncologist. 2021 Mar; 26(3): 242–249.

Published online 2021 Feb 10. doi:10.1002/onco.13685

PMCID: PMC7930415

PMID: 33486852

Julio Delgado,EMA Review of Acalabrutinib for the Treatment of Adult Patients with Chronic Lymphocytic Leukemia (2)1,3 Filip Josephson,2,4 Jorge Camarero,2,5 Blanca Garcia‐Ochoa,2,5 Lucia Lopez‐Anglada,5 Carolina Prieto‐Fernandez,5 Paula B. van Hennik,2,6 Irene Papadouli,1 Christian Gisselbrecht,7 Harald Enzmann,2,8 and Francesco Pignatti1

Author information Article notes Copyright and License information PMC Disclaimer

Abstract

On November 5, 2020, a marketing authorization valid through the European Union (EU) was issued for acalabrutinib monotherapy or acalabrutinib in combination with obinutuzumab (AcalaObi) in adult patients with treatment‐naïve (TN) chronic lymphocytic leukemia (CLL) and also for acalabrutinib monotherapy in adult patients with relapsed or refractory (RR) CLL. Acalabrutinib inhibits the Bruton tyrosine kinase, which plays a significant role in the proliferation and survival of the disease. Acalabrutinib was evaluated in two phase III multicenter randomized trials. The first trial (ACE‐CL‐007) randomly allocated acalabrutinib versus AcalaObi versus chlorambucil plus obinutuzumab (ChlObi) to elderly/unfit patients with TN CLL. The progression‐free survival (PFS), as assessed by an independent review committee, was superior for both the AcalaObi (hazard ratio [HR], 0.1; 95% confidence interval [CI], 0.06–0.17) and acalabrutinib (HR, 0.2; 95% CI, 0.13–0.3) arms compared with the ChlObi arm. The second trial (ACE‐CL‐309) randomly allocated acalabrutinib versus rituximab plus idelalisib or bendamustine to adult patients with RR CLL. Also in this trial, the PFS was significantly longer in the acalabrutinib arm (HR, 0.31; 95% CI, 0.20–0.49). Adverse events for patients receiving acalabrutinib varied across trials, but the most frequent were generally headache, diarrhea, neutropenia, nausea, and infections. The scientific review concluded that the benefit‐risk ratio of acalabrutinib was positive for both indications. This article summarizes the scientific review of the application leading to regulatory approval in the EU.

Implications for Practice

Acalabrutinib was approved in the European Union for the treatment of adult patients with chronic lymphocytic leukemia who have not received treatment before and for those who have received therapy but whose disease did not respond or relapsed afterward. Acalabrutinib resulted in a clinically meaningful and significant lengthening of the time from treatment initiation to further disease relapse or patient's death compared with standard therapy. The overall safety profile was considered acceptable, and the benefit‐risk ratio was determined to be positive.

Keywords: Chronic lymphocytic leukemia, Acalabrutinib, European Medicines Agency, Bruton tyrosine kinase

Short abstract

This article summarizes the EMA scientific review of the application leading to regulatory approval for acalabrutinib for treatment of leukemia.

Introduction

Chronic lymphocytic leukemia (CLL) is the most common form of leukemia in Western countries. The World Health Organization (WHO) describes CLL as a leukemic, lymphocytic lymphoma, being only distinguishable from small lymphocytic lymphoma (SLL) by its leukemic manifestation (≥5,000 monoclonal B cells per microliter in the peripheral blood) [1, 2]. Major progress has been made in the identification of predictive biomarkers, particularly TP53 aberrations (either deletions of chromosome 17p or TP53 mutations) [3, 4].

The treatment of CLL has evolved significantly over the last 2 decades, initially with the advent of fludarabine‐based combinations and later with the addition of monoclonal antibodies targeting CD20. Indeed, fludarabine plus cyclophosphamide plus rituximab (FCR) became the standard of care, whereas bendamustine plus rituximab (BR) and chlorambucil plus obinutuzumab (ChlObi) were recommended for patients who could not tolerate FCR [5, 6, 7, 8, 9, 10, 11, 12, 13]. Patients whose tumors harbor TP53 aberrations have an inferior outcome when treated with these conventional agents [10]. In the last decade, agents targeting Bruton tyrosine kinase (BTK) (ibrutinib), BCL2 (venetoclax) or PI3Kδ (idelalisib) have shown relevant efficacy in patients with CLL, particularly those with TP53 aberrations, and are all approved in the European Union (EU) [12, 14, 15, 16]. Specifically, ibrutinib is approved (a) in monotherapy or in combination with rituximab or obinutuzumab for the treatment of treatment‐naïve (TN) CLL and (b) in monotherapy or in combination with BR for the treatment of relapsed or refractory (RR) CLL. Venetoclax is approved (a) in combination with obinutuzumab for the treatment of TN CLL, (b) in combination with rituximab for the treatment of RR CLL, and (c) in monotherapy for the treatment of patients with TP53 aberrations failing ibrutinib/idelalisib or in those without TP53 aberrations failing both chemoimmunotherapy and ibrutinib/idelalisib. Finally, idelalisib is approved (a) in combination with rituximab or ofatumumab for the treatment of RR CLL and (b) as first‐line therapy in patients with TP53 aberrations who are ineligible for other therapies.

On October 14, 2019, AstraZeneca AB, Sweden, applied for a marketing authorization via the European Medicines Agency (EMA) centralized procedure for acalabrutinib (trade name Calquence) for the treatment of CLL. Acalabrutinib had been designated orphan medicine by the European Commission in March 2016. To qualify for orphan designation, a medicine must be intended for the treatment, prevention, or diagnosis of a life‐threatening or chronically debilitating disease; the prevalence of the condition in the EU must not be more than 5 in 10,000; and the medicine must be of significant benefit to those affected by the condition.

The review was conducted by the Committee for Medicinal Products for Human Use (CHMP), and a positive opinion was issued on July 23, 2020. The approved indication in the EU is as follows: “Calquence monotherapy or in combination with obinutuzumab is indicated for treatment of adult patients with previously untreated CLL. Calquence monotherapy is indicated for treatment of adult patients with CLL who have received at least one prior therapy.” This article summarizes the scientific review of the application leading to the regulatory approval of acalabrutinib in the EU.

Nonclinical Aspects

Acalabrutinib is a selective inhibitor of BTK, a Tec family tyrosine kinase normally expressed in B cells, myeloid cells, mast cells, and platelets. Targeted inhibition of BTK with ibrutinib and acalabrutinib leads to antitumor activity in various B‐cell malignancies, including CLL [17]. in vitro assays showed that acalabrutinib inhibits BTK, ERBB4, and BLK, whereas ibrutinib displayed inhibitory activity toward eight out of nine kinases. In comparison with ibrutinib, acalabrutinib possessed reduced activity for TEC, EGFR, and ITK and no activity on Src‐family kinases. As a result, ibrutinib inhibited T‐cell activation, EGFR activation of epidermal cells, and natural killer (NK)‐cell killing, whereas acalabrutinib showed no or little activity [18].

Pharmacology

The pharmaco*kinetics of acalabrutinib were studied in mice, rats, dogs, monkeys, and human tissues. The single‐dose pharmaco*kinetic properties of acalabrutinib were characterized by high clearance and short half‐life after oral administration in all nonclinical test species. The drug was also evaluated in a phase I–II multicenter, open‐label, dose‐escalation clinical trial in patients with RR CLL (ACE‐CL‐001) [19, 20]. In this trial, the drug was rapidly absorbed and eliminated, with a mean half‐life of approximately 1 hour and no plasma accumulation. BTK occupancy was complete (99%–100%) 4 hours after dosing but decreased to 87%–95% before subsequent dose administration with once‐daily dosing. Twice‐daily dosing, on the other hand, improved BTK occupancy before administration to 97%, with complete loss of BTK phosphorylation at all time points evaluated [19]. The high selectivity of acalabrutinib was confirmed by the sustained platelet–vessel wall interactions in platelets isolated from patients receiving acalabrutinib (compared with a significant reduction in platelets isolated from patients receiving ibrutinib), as well as intact NK cell–mediated cytotoxicity in peripheral blood. Repeat‐dose toxicity was evaluated in mice, rats, and dogs. The drug was well tolerated at lower doses, although some hematology findings, mild liver toxicity, and lower spleen weight were observed in acalabrutinib‐treated animals.

Trial Design

The application was based on the pivotal phase III clinical trials ACE‐CL‐007 and ACE‐CL‐309 [21, 22]. Acalabrutinib had been previously evaluated as monotherapy [19, 20] or in combination with obinutuzumab [23] in two phase Ib–II clinical trials. In these trials, the recommended phase II dose was established at 100 mg twice daily, whereas obinutuzumab was added at the currently approved dose. No formal dose escalation was performed for the combined therapy.

ACE‐CL‐007

ACE‐CL‐007 was a randomized, multicenter, open‐label study of ChlObi (arm A), acalabrutinib in combination with obinutuzumab (AcalaObi) (arm B), and acalabrutinib monotherapy (arm C) in patients with TN CLL. Only unfit (creatinine clearance of 30–69 mL/min, or cumulative illness rating scale‐geriatric > 6) or elderly (≥65 years) patients were allowed in this trial. Treatment was administered in 28‐day cycles. In arms B and C, oral acalabrutinib was administered (100 mg twice daily) until progressive disease (PD) or unacceptable toxicity. In arm A, intravenous obinutuzumab was given on day 1 (100 mg), day 2 (900 mg), day 8 (1,000 mg), and day 15 (1,000 mg) of cycle 1 and on day 1 (1,000 mg) of cycles 2–6. In arm B, obinutuzumab was administered following the same schedule but with 1‐month delay (on cycles 2–7). In arm A, oral chlorambucil was administered (0.5 mg/kg) on days 1 and 15 of each of these six cycles. Patients allocated to arm A were able to cross over to receive acalabrutinib in case of PD.

ACE‐CL‐309

ACE‐CL‐309 was a randomized, multicenter, open‐label study of acalabrutinib (arm A) versus idelalisib plus rituximab (IR) or BR (arm B) in patients with RR CLL. Of note, the choice between IR or BR was at the discretion of the investigator. The study enrolled subjects with at least one prior systemic therapy, but prior BCL2 or BTK/PI3K inhibitors were not allowed. Treatment was administered in 28‐day cycles. In arm A, oral acalabrutinib (100 mg twice daily) was administered until PD or unacceptable toxicity. In arm B, oral idelalisib (150 mg twice daily) was administered until PD or unacceptable toxicity in combination with intravenous rituximab (375 mg/m2 on day 1 of the first cycle, followed by 500 mg/m2 every 2 weeks for four doses and then every 4 weeks for three doses for a total of eight infusions). Alternatively, patients in arm B could receive intravenous bendamustine 70 mg/m2 on days 1 and 2 of each cycle in combination with intravenous rituximab (375 mg/m2 on day 1 of the first cycle, 500 mg/m2 on day 1 or cycles 2–6). Patients in arm B with confirmed PD could cross over to arm A.

In both trials, patients with prior history of significant cardiovascular disease, stroke, intracranial hemorrhage, bleeding diathesis, or requiring anticoagulation with vitamin K antagonists were excluded. In both trials, the primary endpoint was progression‐free survival (PFS) as assessed by an independent review committee (IRC). In trial ACE‐CL‐007, two comparisons were planned: the first between arm A and arm B (primary endpoint) and the second between arm A and arm C (key secondary endpoint).

Clinical Efficacy

ACE‐CL‐007

ACE‐CL‐007 randomized 535 subjects: 177 to arm A, 179 to arm B, and 179 to arm C. With a median follow‐up of 28 months, the primary endpoint showed a hazard ratio (HR) of 0.10 (95% confidence interval [CI], 0.06–0.17) in favor of arm B (AcalaObi). Moreover, the median IRC‐confirmed PFS was 22.6 months for arm A (ChlObi) (95% CI, 20.2–27.6), whereas it had not been reached for arm B (Fig. ​(Fig.1;1; Table ​Table1).1). Subgroup analyses consistently favored arm B, including patients with or without TP53 aberrations. The HR for the key secondary endpoint was 0.20 (95% CI, 0.13–0.30) in favor of arm C (acalabrutinib monotherapy). Subgroup analyses consistently favored arm C (acalabrutinib monotherapy) over arm A (ChlObi).

Open in a separate window

Figure 1

Kaplan‐Meier plots for progression‐free survival by independent review committee assessment (primary and key secondary endpoints) in study ACE‐CL‐007.Abbreviations: Acala, acalabrutinib; Obin, obinutuzumab.

Table 1

Key favorable and unfavorable of acalabrutinib therapy for adult patients with chronic lymphocytic leukemia (study ACE‐CL‐007 cutoff date: January 2019)

Effect (previously untreated disease)Arm B: acalabrutinib + obinutuzumabArm C: acalabrutinibArm A (control): chlorambucil + obinutuzumabUncertainties, strength of evidence
Favorable effects
PFS by IRCHR, 0.1 (95% CI, 0.06–0.17); p < .0001HR, 0.2 (95% CI, 0.13–0.3); p < .0001

Median follow‐up, 28 mo

Control arm:

Event rate, 52%

Median PFS, 22.6 mo

ORR by IRC, %93.9 (95% CI, 89.3–96.5)85.5 (95% CI, 79.6–89.9)78.5 (71.9–83.9)
TTNTHR, 0.14 (95% CI, 0.08–0.26)HR, 0.24 (95% CI, 0.15–0.40)Median not reached in any arm
Unfavorable effects, %
Grade ≥3 TEAEs70.249.769.8
Grade ≥3 SAEs32.629.619.5
Cardiac events14.014.07.7
Hypertension7.34.53.6
Neutropenia31.510.645.0
Bleeding42.739.111.8
Hepatotoxicity9.02.24.7
Infections69.165.443.8
Pneumonitis0.61.10.6
SPMs10.78.43.6

Open in a separate window

Abbreviations: CI, confidence interval; HR, hazard ratio; IRC, independent review committee; ORR, overall response rate; PFS, progression‐free survival; SAE, severe adverse event; SPM, secondary primary malignancy; TEAE, treatment‐emergent adverse event; TTNT, time to next therapy.

Regarding other secondary endpoints, the IRC‐assessed overall response rate (ORR) was 78.5% (95% CI, 71.9–83.9), 93.9% (95% CI, 89.3–96.5), and 85.5% (95% CI, 79.6–89.9) for arms A, B, and C, respectively; time to next therapy (TTNT) was significantly prolonged for arm B (HR, 0.14; 95% CI, 0.08–0.26) and arm C (HR, 0.24; 95% CI, 0.15–0.40) compared with arm A; and there were no significant differences in terms of overall survival (OS), although the data were immature and crossover was allowed (25% of patients from arm A crossed over to acalabrutinib therapy).

ACE‐CL‐309

ACE‐CL‐309 randomized 310 subjects: 155 to arm A (acalabrutinib) and 155 to arm B (119 received IR and 36 received BR). With a median follow‐up of 16 months, the primary endpoint showed an HR of 0.31 (95% CI, 0.20–0.49) in favor of arm A (acalabrutinib). Moreover, the median IRC‐confirmed PFS was 16.5 months for arm B (95% CI, 14.0–17.1), whereas it had not been reached for arm A. Subgroup analyses consistently favored arm A (acalabrutinib), including patients with or without TP53 aberrations. These results were updated during the procedure, and at a median follow‐up of 22 months the HR for PFS was 0.27 (95% CI, 0.18–0.40) (Fig. ​(Fig.2;2; Table ​Table22).

Open in a separate window

Figure 2

Kaplan‐Meier plot for progression‐free survival by investigator assessment (primary endpoint) in study ACE‐CL‐309. Arm A corresponds to treatment with acalabrutinib, and arm B corresponds to treatment with idelalisib plus rituximab or bendamustine plus rituximab.Abbreviations: CI, confidence interval; HR, hazard ratio.

Table 2

Key favorable and unfavorable of acalabrutinib therapy for adult patients with chronic lymphocytic leukemia (study ACE‐CL‐309, cutoff date: January 2019)

Effect (relapsed or refractory patients)Arm A: acalabrutinibArm B: controlUncertainties, strength of evidence
Idelalisib + rituximabBendamustine + rituximab
Favorable effects
PFS by IRCHR, 0.31 (95% CI, 0.20–0.49); p < .0001

Median follow‐up, 16 mo

Control arm:

Event rate, 44%

Median PFS, 16.5 mo

ORR by IRC, %81.3 (95% CI, 74.4–86.6)75.5 (95% CI, 68.1–81.6)
Median DOR by IRCNR13.6 mo
Unfavorable effects, %
Grade ≥3 TEAEs49.489.848.6
Grade ≥3 SAEs26.650.825.7
Cardiac events13.07.68.6
Hypertension3.24.20
Neutropenia21.450.837.1
Bleeding26.07.65.7
Hepatotoxicity4.528.08.6
Infections56.565.348.6
Pneumonitis1.96.80
SPMs11.72.52.9

Open in a separate window

Abbreviations: CI, confidence interval; DOR, duration of response; HR, hazard ratio; IRC, independent review committee; NR, not reached; ORR, overall response rate; PFS, progression‐free survival; SAE, severe adverse event; SPM, secondary primary malignancy; TEAE, treatment‐emergent adverse event.

Regarding other secondary endpoints, there were no statistical differences in ORR, and median TTNT had not been reached for any of the study arms, but HR was 0.35 (95% CI, 0.21–0.58) favoring arm A over arm B. There were no significant differences in OS, but the data were immature and crossover was allowed (23% of patients from arm B crossed over to acalabrutinib therapy).

Clinical Safety

The safety database comprised 1,040 subjects from nine studies (Mono HemMalig) in patients receiving acalabrutinib monotherapy (ACE‐CL‐309, ACE‐CL‐007, 15‐H‐0016, ACE‐CL‐001, ACE‐LY‐002, ACE‐LY‐003, ACE‐LY‐004, ACE‐MY‐001, and ACE‐WM‐001) and two studies (ACE‐CL‐007, ACE‐CL‐003) in patients with CLL receiving AcalaObi (ComboCLL, n = 223). Median time of follow‐up was 24.6 months for the Mono HemMalig pooled population and 29.8 months for the ComboCLL pooled population [19, 20, 21, 22, 23, 24, 25, 26].

ACE‐CL‐007

In ACE‐CL‐007, evaluation of adverse events (AEs) is complicated by the fact that the maximum time on therapy was 6 months for arm A but until PD or unacceptable toxicity for arms B and C. The median relative dose intensity was >98% in both acalabrutinib arms (arms B and C). The most frequent treatment‐emergent AEs (TEAEs) in patients from arm A were neutropenia (45.0%), infusion‐related reactions (39.6%), nausea (31.4%), diarrhea (21.3%), and pyrexia (20.7%). Most of these TEAEs were grade 1–2 apart from neutropenia (grade ≥3 in 41.4%). In patients from arm B, the most frequent TEAEs were headache (39.9%), diarrhea (38.8%), neutropenia (31.5%), fatigue (28.1%), contusion (23.6%), arthralgia (21.9%), cough (21.9%), upper respiratory tract infection (21.3%), and nausea (20.2%). Most TEAEs were grade 1–2 except for neutropenia (grade ≥3 in 29.8%). In patients from arm C, the most frequent TEAEs were headache (36.9%), diarrhea (34.6%), and nausea (22.3%). Only two grade 3 TEAEs were recorded in arm C (one case of headache and one case of diarrhea), with no grade ≥4 TEAEs in this arm. Second primary malignancies (SPMs) were observed in 3.6% of patients from arm A, 10.7% of patients from arm B, and 8.4% of patients from arm C, with basal cell cutaneous carcinoma being the most frequent histology. Atrial fibrillation (any grade) was recorded in 0.6%, 3.4%, and 3.9% of patients from arms A, B, and C, respectively, whereas grade ≥3 bleeding episodes were observed in 0%, 1.7%, and 1.7% patients from arms A, B, and C, respectively.

Deaths because of AEs were documented in 10 (5.9%), 4 (2.2%), and 6 (3.4%) patients from arms A, B, and C, respectively. Grade ≥3 severe AEs (SAEs) were observed in 19%, 32%, and 29% of patients from arms A, B, and C, respectively. Several events of clinical interest were also investigated based on prior studies with acalabrutinib and the approved BTK inhibitor ibrutinib and are displayed in Table ​Table11.

ACE‐CL‐309

In ACE‐CL‐309, the drug exposure was also different among cohorts. The actual median time on treatment was 15.7 months for acalabrutinib (arm A), 11.5 for IR (arm B), and 5.6 months for BR (arm B). The median relative dose intensity of acalabrutinib was 99.5%. The most common TEAEs in patients from arm A were headache (22.1%), neutropenia (19.5%), diarrhea (18.2%), anemia (14.9%), cough (14.9%), upper respiratory infection (14.3%), pyrexia (12.3%), thrombocytopenia (11.0%), pneumonia (10.4%), and respiratory tract infection (10.4%). Only one patient had a grade ≥3 TEAE (febrile neutropenia). In patients from arm B (IR group), the most common TEAEs were diarrhea (46.6%), neutropenia (44.9%), pyrexia (17.8%), cough (15.3%), upper respiratory tract infection (14.4%), thrombocytopenia (13.6%), rash (13.6%), nausea (12.7%), pneumonia (11.9%), and increased alanine aminotransferase (11.9%). Most TEAEs were grade 1–2. In patients from arm B (BR group), the most common TEAEs were neutropenia (34.3%), fatigue (22.9%), infusion‐related reaction (22.9%), nausea (20.0%), pyrexia (17.1%), constipation (14.3%), diarrhea (14.3%), thrombocytopenia (14.3%), anemia (11.4%), and upper respiratory tract infection (11.4%). Most TEAEs were grade 1–2. SPMs were documented in 11.7% of patients treated with acalabrutinib compared with 2.5% and 2.9% of patients treated with IR and BR, respectively. Atrial fibrillation (any grade) was recorded in 5.2%, 2.5%, and 2.9% of patients from arms A, B (IR group), and B (BR group), respectively. Grade ≥3 bleeding episodes occurred in 1.9%, 2.5%, and 2.9% of patients treated with acalabrutinib, IR, and BR, respectively.

Deaths because of AEs were observed in 15 (9.7%) and 18 (11.8%) patients from arms A and B, respectively, including three subjects who had already crossed over to acalabrutinib therapy. SAEs occurred in 28.6%, 55.9%, and 25.7% of patients who received acalabrutinib, IR, and BR, respectively, with most SAEs being of grade 3 or higher.

Benefit‐Risk Assessment

For patients with TN CLL, the PFS benefit observed in both experimental arms (arm B, AcalaObi; and arm C, acalabrutinib) compared with the control arm (arm A, ChlObi) was considered robust and clinically relevant. Moreover, the safety profile appeared similar that of the approved BTK inhibitor ibrutinib, including comparable rates of cardiovascular events (e.g., atrial fibrillation), diarrhea, cytopenia, and hemorrhage [14, 27, 28]. The potential impact of the experimental arms on OS could not be fully evaluated because of differences in drug exposure (until PD or unacceptable toxicity vs. fixed duration) and crossover. Regarding both experimental arms (B and C), the trial was not powered for that comparison, but point estimates for ORR and PFS numerically favored the combination arm. Toxicity was somewhat higher in patients receiving AcalaObi (higher frequency and severity of AEs), although this did not translate in a higher proportion of permanent discontinuations or a decreased relative dose intensity in the AcalaObi arm. The benefit‐risk ratio was considered positive for both experimental regimens, and it was therefore considered appropriate to approve both. Of note, the study ACE‐CL‐007 only enrolled older or comorbid patients, but, based on the precedent set for ibrutinib, the extrapolation of efficacy to younger or fitter patients was considered acceptable, and safety was not expected to be worse.

The use of ChlObi as control therapy was initially questioned for patients with TP53 aberrations. At the time of study initiation, European Society for Medical Oncology guidelines recommended a BCR inhibitor (e.g., ibrutinib) for patients with this molecular profile [29]. However, the applicant further justified the choice of comparator alluding to historical reasons, which was acknowledged and accepted. Still, data from the ACE‐CL‐007 study revealed a similarly favorable PFS for both experimental arms irrespective of TP53 status, with ORR as high as 83%–84% in patients with TP53 aberrations compared with 56% for the control arm.

For patients with RR CLL, the PFS benefit observed in the experimental arm (acalabrutinib) compared with the control arm (IR/BR) was deemed robust and clinically relevant, with retained activity in patients with TP53 aberrations. No major unexpected AEs were observed in the experimental arm. The major uncertainty was low maturity at data cutoff, with a PFS event rate of only 44% in the control arm. An updated analysis (per investigator assessment) based on 6 months of further follow‐up was provided, and with a PFS event rate of 58% in the control arm, the point estimates remained essentially unchanged. The final clinical study reports for studies ACE‐CL‐007 (including data on time to second subsequent therapy) and ACE‐CL‐309 are expected to be reviewed by the CHMP as postapproval data.

Regarding safety, final study reports from pivotal studies and safety updates from study D8220C00008, a phase IIIb, multicenter, open‐label, single‐arm study of acalabrutinib in subjects with CLL will be reviewed postauthorization. Moreover, the ongoing phase III randomized controlled trial ACE‐CL‐006 is currently comparing the efficacy and safety of acalabrutinib versus ibrutinib in patients with high‐risk RR CLL. Cardiac events beyond atrial fibrillation occurred at a higher rate in the acalabrutinib‐containing arms compared with comparator arms in both pivotal studies and must be further characterized. Data from ongoing studies, including a cohort of study D8220C00008, will provide information on the use of acalabrutinib in patients with moderate or severe cardiac impairment. Moreover, the inherent risk of patients with CLL to develop SPMs did not allow the CHMP to fully characterize the acalabrutinib‐related risk of SPMs, which are presently included as an important identified risk in the safety specifications. SPMs will also be further characterized in upcoming postauthorization studies.

Patients receiving anticoagulation were not included in these studies because of a high bleeding risk, and the same applied to proton pump inhibitors or strong CYP3A inducers or inhibitors to prevent pharmacologic interactions. Accordingly, the recommendation to avoid the coadministration of acalabrutinib and these agents was added to the Summary of Product Characteristics (SmPC) and endorsed by the CHMP.

The proposed indication for Calquence in the RR setting includes patients previously treated with ibrutinib or venetoclax, who were excluded from the ACE‐CL‐309 trial. The applicant provided external data reasonably supporting the use of BTK inhibitors in subjects previously treated with venetoclax and the use of acalabrutinib in patients intolerant (but not resistant) to ibrutinib. This is mechanistically expected and was added to the SmPC. Finally, the applicant proposed to add the term SLL to the indication, which was refused by the CHMP on the grounds that CLL and SLL are considered the same disease by the WHO [1].

Conclusion

Based on the review of data on quality, safety, and efficacy, the EMA CHMP concluded by consensus that the risk‐benefit balance of acalabrutinib monotherapy and acalabrutinib in combination with obinutuzumab was favorable for the treatment of adult patients with previously untreated CLL, and hence recommended the granting of the marketing authorization. Moreover, the risk‐benefit balance of acalabrutinib monotherapy was also favorable for the treatment of adult patients with relapsed or refractory CLL, also recommending the granting of the marketing authorization.

Author Contributions

Data analysis and interpretation: Filip Josephson, Jorge Camarero, Blanca Garcia‐Ochoa, Lucia Lopez‐Anglada, Carolina Prieto‐Fernandez, Paula B. van Hennik, Harald Enzmann

Manuscript writing: Julio Delgado, Filip Josephson, Jorge Camarero, Blanca Garcia‐Ochoa, Paula B. van Hennik, Irene Papadouli, Christian Gisselbrecht, Harald Enzmann, Francesco Pignatti

Final approval of manuscript: Julio Delgado, Filip Josephson, Jorge Camarero, Blanca Garcia‐Ochoa, Lucia Lopez‐Anglada, Carolina Prieto‐Fernandez, Paula B. van Hennik, Irene Papadouli, Christian Gisselbrecht, Harald Enzmann, Francesco Pignatti

Disclosures

The authors indicated no financial relationships.

Acknowledgments

The scientific assessment summarized in this report is based on important contributions from the rapporteur and corapporteur assessment teams, Committee for Medicinal Products for Human Use members, and additional experts after the application for a marketing authorization from the company. This publication is a summary of the European Public Assessment Report, the Summary of Product Characteristics, and other product data as published on the European Medicines Agency (EMA) Web site (https://www.ema.europa.eu/en/medicines). For the most current information on this marketing authorization, please refer to the EMA Web site. The views expressed in this article are the personal views of the author(s) and may not be understood or quoted as being made on behalf of or reflecting the position of the regulatory agency/agencies or organizations with which the author(s) is/are employed/affiliated.

Notes

Disclosures of potential conflicts of interest may be found at the end of this article.

No part of this article may be reproduced, stored, or transmitted in any form or for any means without the prior permission in writing from the copyright holder. For information on purchasing reprints contact moc.yeliw@stnirperlaicremmoc. For permission information contact moc.yeliw@snoissimrep.

Footnotes

For Further Reading: Paul J. Hampel, Timothy G. Call, Kari G. Rabe et al. Disease Flare During Temporary Interruption of Ibrutinib Therapy in Patients with Chronic Lymphocytic Leukemia. The Oncologist 2020;25:974–980.

Implications for Practice: Ibrutinib is a very effective treatment for chronic lymphocytic leukemia (CLL) but needs to be taken continuously. Side effects, such as increased bleeding risk with procedures, require temporary interruptions in this continuous treatment. Rapid CLL progression following ibrutinib discontinuation has been increasingly recognized. This study demonstrates that similar flares in disease signs or symptoms may occur during ibrutinib holds as well. Importantly, management with restarting ibrutinib led to quick clinical improvement. Awareness of this phenomenon among clinicians is critical to avoid associated patient morbidity and premature cessation of effective treatment with ibrutinib if the flare is misidentified as true progression of disease.

References

1. Swerdlow SH, Campo E, Pileri SA et al. The 2016 revision of the World Health Organization classification of lymphoid neoplasms. Blood2016;127:2375–2390. [PMC free article] [PubMed] [Google Scholar]

2. Sant M, Allemani C, Tereanu C et al. Incidence of hematologic malignancies in Europe by morphologic subtype: Results of the HAEMACARE project. Blood2010;116:3724–3734. [PubMed] [Google Scholar]

3. Hallek M.Chronic lymphocytic leukemia: 2020 update on diagnosis, risk stratification and treatment. Am J Hematol2019;94:1266–1287. [PubMed] [Google Scholar]

4. Hallek M, Cheson BD, Catovsky D et al. iwCLL guidelines for diagnosis, indications for treatment, response assessment, and supportive management of CLL. Blood2018;131:2745–2760. [PubMed] [Google Scholar]

5. Catovsky D, Richards S, Matutes E et al. Assessment of fludarabine plus cyclophosphamide for patients with chronic lymphocytic leukaemia (the LRF CLL4 Trial): A randomised controlled trial. Lancet2007;370:230–239. [PubMed] [Google Scholar]

6. Flinn IW, Neuberg DS, Grever MR et al. Phase III trial of fludarabine plus cyclophosphamide compared with fludarabine for patients with previously untreated chronic lymphocytic leukemia: US Intergroup Trial E2997. J Clin Oncol2007;25:793–798. [PubMed] [Google Scholar]

7. Eichhorst BF, Busch R, Hopfinger G et al. Fludarabine plus cyclophosphamide versus fludarabine alone in first‐line therapy of younger patients with chronic lymphocytic leukemia. Blood2006;107:885–891. [PubMed] [Google Scholar]

8. Keating MJ, O'Brien S, Albitar M et al. Early results of a chemoimmunotherapy regimen of fludarabine, cyclophosphamide, and rituximab as initial therapy for chronic lymphocytic leukemia. J Clin Oncol2005;23:4079–4088. [PubMed] [Google Scholar]

9. Wierda W, O'Brien S, Wen S et al. Chemoimmunotherapy with fludarabine, cyclophosphamide, and rituximab for relapsed and refractory chronic lymphocytic leukemia. J Clin Oncol2005;23:4070–4078. [PubMed] [Google Scholar]

10. Hallek M, Fischer K, Fingerle‐Rowson G et al. Addition of rituximab to fludarabine and cyclophosphamide in patients with chronic lymphocytic leukaemia: A randomised, open‐label, phase 3 trial. Lancet2010;376:1164–1174. [PubMed] [Google Scholar]

11. Robak T, Dmoszynska A, Solal‐Celigny P et al. Rituximab plus fludarabine and cyclophosphamide prolongs progression‐free survival compared with fludarabine and cyclophosphamide alone in previously treated chronic lymphocytic leukemia. J Clin Oncol2010;28:1756–1765. [PubMed] [Google Scholar]

12. Goede V, Fischer K, Busch R et al. Obinutuzumab plus chlorambucil in patients with CLL and coexisting conditions. N Engl J Med2014;370:1101–1110. [PubMed] [Google Scholar]

13. Eichhorst B, Fink AM, Bahlo J et al. First‐line chemoimmunotherapy with bendamustine and rituximab versus fludarabine, cyclophosphamide, and rituximab in patients with advanced chronic lymphocytic leukaemia (CLL10): An international, open‐label, randomised, phase 3, non‐inferiority trial. Lancet Oncol2016;17:928–942. [PubMed] [Google Scholar]

14. Byrd JC, Brown JR, O'Brien S et al. Ibrutinib versus ofatumumab in previously treated chronic lymphoid leukemia. N Engl J Med2014;371:213–223. [PMC free article] [PubMed] [Google Scholar]

15. Roberts AW, Davids MS, Pagel JM et al. Targeting BCL2 with venetoclax in relapsed chronic lymphocytic leukemia. N Engl J Med2016;374:311–322. [PMC free article] [PubMed] [Google Scholar]

16. Furman RR, Sharman JP, Coutre SE et al. Idelalisib and rituximab in relapsed chronic lymphocytic leukemia. N Engl J Med2014;370:997–1007. [PMC free article] [PubMed] [Google Scholar]

17. Wiestner A. Emerging role of kinase‐targeted strategies in chronic lymphocytic leukemia. Blood2012;120:4684–4691. [PMC free article] [PubMed] [Google Scholar]

18. Barf T, Covey T, Izumi R et al. Acalabrutinib (ACP‐196): A covalent bruton tyrosine kinase inhibitor with a differentiated selectivity and in vivo potency profile. J Pharmacol Exp Ther2017;363:240–252. [PubMed] [Google Scholar]

19. Byrd JC, Harrington B, O'Brien S et al. Acalabrutinib (ACP‐196) in relapsed chronic lymphocytic leukemia. N Engl J Med2016;374:323–332. [PMC free article] [PubMed] [Google Scholar]

20. Byrd JC, Wierda WG, Schuh A et al. Acalabrutinib monotherapy in patients with relapsed/refractory chronic lymphocytic leukemia: Updated phase 2 results. Blood2020;135:1204–1213. [PMC free article] [PubMed] [Google Scholar]

21. Sharman JP, Egyed M, Jurczak W et al. Acalabrutinib with or without obinutuzumab versus chlorambucil and obinutuzmab for treatment‐naive chronic lymphocytic leukaemia (ELEVATE TN): A randomised, controlled, phase 3 trial. Lancet2020;395:1278–1291. [PMC free article] [PubMed] [Google Scholar]

22. Ghia P, Pluta A, Wach M et al. ASCEND: Phase III, randomized trial of acalabrutinib versus idelalisib plus rituximab or bendamustine plus rituximab in relapsed or refractory chronic lymphocytic leukemia. J Clin Oncol2020;38:2849–2861. [PubMed] [Google Scholar]

23. Woyach JA, Blachly JS, Rogers KA et al. Acalabrutinib plus obinutuzumab in treatment‐naive and relapsed/refractory chronic lymphocytic leukemia. Cancer Discov2020;10:394–405. [PMC free article] [PubMed] [Google Scholar]

24. Sun C, Nierman P, Kendall EK et al. Clinical and biological implications of target occupancy in CLL treated with the BTK inhibitor acalabrutinib. Blood2020;136:93–105. [PMC free article] [PubMed] [Google Scholar]

25. Owen RG, McCarthy H, Rule S et al. Acalabrutinib monotherapy in patients with Waldenstrom macroglobulinemia: A single‐arm, multicentre, phase 2 study. Lancet Haematol2020;7:e112–e121. [PubMed] [Google Scholar]

26. Wang M, Rule S, Zinzani PL et al. Acalabrutinib in relapsed or refractory mantle cell lymphoma (ACE‐LY‐004): A single‐arm, multicentre, phase 2 trial. Lancet2018;391:659–667. [PMC free article] [PubMed] [Google Scholar]

27. Burger JA, Tedeschi A, Barr PM et al. Ibrutinib as initial therapy for patients with chronic lymphocytic leukemia. N Engl J Med2015;373:2425–2437. [PMC free article] [PubMed] [Google Scholar]

28. Moreno C, Greil R, Demirkan F et al. Ibrutinib plus obinutuzumab versus chlorambucil plus obinutuzumab in first‐line treatment of chronic lymphocytic leukaemia (iLLUMINATE): A multicentre, randomised, open‐label, phase 3 trial. Lancet Oncol2019;20:43–56. [PubMed] [Google Scholar]

29. Eichhorst B, Robak T, Montserrat E et al. Chronic lymphocytic leukaemia: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow‐up. Ann Oncol2015;26(suppl 5):v78–v84. [PubMed] [Google Scholar]

Articles from The Oncologist are provided here courtesy of Oxford University Press

EMA Review of Acalabrutinib for the Treatment of Adult Patients with Chronic Lymphocytic Leukemia (2024)

References

Top Articles
Latest Posts
Article information

Author: The Hon. Margery Christiansen

Last Updated:

Views: 5925

Rating: 5 / 5 (70 voted)

Reviews: 93% of readers found this page helpful

Author information

Name: The Hon. Margery Christiansen

Birthday: 2000-07-07

Address: 5050 Breitenberg Knoll, New Robert, MI 45409

Phone: +2556892639372

Job: Investor Mining Engineer

Hobby: Sketching, Cosplaying, Glassblowing, Genealogy, Crocheting, Archery, Skateboarding

Introduction: My name is The Hon. Margery Christiansen, I am a bright, adorable, precious, inexpensive, gorgeous, comfortable, happy person who loves writing and wants to share my knowledge and understanding with you.