Original Article

Split Viewer

Blood Res 2023; 58(4):

Published online December 31, 2023

https://doi.org/10.5045/br.2023.2023208

© The Korean Society of Hematology

A retrospective analysis of ibrutinib outcomes in relapsed or refractory mantle cell lymphoma

Yong-Pyo Lee1#, Ye Ji Jung2#, Junhun Cho3, Young Hyeh Ko3, Won Seog Kim2,4, Seok Jin Kim2,4, Sang Eun Yoon2

1Division of Hematology-Oncology, Department of Internal Medicine, Chungbuk National University Hospital, Cheongju, Division of Hematology-Oncology, Departments of 2Medicine, 3Pathology, Samsung Medical Center, Sungkyunkwan University School of Medicine, 4Health Sciences and Technology, Samsung Advanced Institute for Health Sciences and Technology, Sungkyunkwan University, Seoul, Korea

Correspondence to : Sang Eun Yoon, M.D., Ph.D.
Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, 81 Irwon-ro, Gangnam-gu, Seoul 06351, Korea
E-mail: starload0326@gmail.com

#These authors contributed equally to this work.

*This study was supported by a grant from the Korea Health Technology R&D Project through the Korea Health Industry Development Institute (KHIDI), funded by the Ministry of Health and Welfare, Republic of Korea (grant number: HI22C0999). This study was also supported by grants from the Korea Health Technology R&D Project through the Korea Health Industry Development Institute, funded by the Ministry of Health & Welfare, Republic of Korea (HR20C0025); a National Research Foundation of Korea grant funded by the Korean government (2022R1F1A1064058); and the Bio&Medical Technology Development Program of the National Research Foundation (NRF) funded by the Korean government (MSIT) (No. RS-2023-00222838).

Received: October 26, 2023; Revised: November 27, 2023; Accepted: December 8, 2023

This is an Open Access article distributed under the terms of the Creative Commons Attribution Non-Commercial License (http://creativecommons.org/licenses/by-nc/4.0) which permits unrestricted non-commercial use, distribution, and reproduction in any medium, provided the original work is properly cited.

Background
While treatment strategies for mantle cell lymphoma (MCL) have evolved, patients often experience disease progression and require additional treatment therapies. Ibrutinib presents a promising option for relapsed or refractory MCL (RR-MCL). This study investigated real-world treatment outcomes of ibrutinib in patients with RR-MCL.
Methods
A single-center retrospective analysis investigated clinical characteristics and survival outcomes of patients with RR-MCL, treated with ibrutinib.
Results
Forty-two patients were included, with 16 received rituximab and bendamustine, and 26 receiving anthracycline-based regimens as front-line treatment. During a median follow-up of 46.0 months, the response rate to ibrutinib was 69%, with 12 CRs and 8 partial responses. Disease progression (54.8%) and adverse events (11.9%) were the primary reasons for discontinuation. Median progression-free survival (PFS) and overall survival (OS) were approximately 16.4 and 50.1 months, respectively. Patients older than 70 years (P=0.044 and P=0.006), those with splenomegaly (P=0.022 and P=0.006), and those with a high-risk simplified Mantle Cell Lymphoma International Prognostic Index (sMIPI) (P<0.001 and P<0.001) exhibited siginificantly inferior PFS and OS. Notably, patients with a high-risk sMIPI relapsed earlier. Post-ibrutinib treatment yilded an OS of 12.2 months, while clinical trial participants demonstrated superior survival compared to those receiving chemotherapy alone.
Conclusion
This study underscores the importance of considering patient characteristics before administering ibrutinib as salvage therapy. Early relapse was associated with poor outcomes, highlighting the need for novel therapeutic strategies.

Mantle cell lymphoma (MCL) is a distinct subtype of non-Hodgkin’s, accounting for approximately 2% of B-cell malignancies [1, 2]. According to an understanding of the developmental etiology of MCL, deregulation of B-cell receptor (BCR) signaling and Bruton tyrosine kinase (BTK) permits continuous B-cell proliferation, leading to MCL development in the pre-germinal center. BTK inhibitors block BCR signaling propagation, related to B-cell proliferation, and allow cancer cell apoptosis by ultimately reducing nuclear factor-κB pathway activity [3]. Thus, BTK is a key factor in BCR signaling and a therapeutic target for MCL. Several covalent (ibrutinib, acalabrutinib, and zanubrutinib) and non-covalent (pirtobrutinib) BTK inhibitors have been developed [4].

Among the various BTK inhibitors, ibrutinib, the first oral selective BTK inhibitor, has a favorable objective response rate (ORR) of 65–70% with a complete response (CR) rate of 19% and progression-free survival (PFS) of 12–15 months for relapsed/refractory MCL (RR-MCL) patients, who received more than two treatments prior [5-15]. Previous studies suggested that the response to ibrutinib depended on the simplified Mantle Cell Lymphoma Inter-national Prognostic Index (sMIPI), which is calculated based on age, performance status, lactate dehydrogenase (LDH), and white blood cell (WBC) count. In addition to the sMIPI, other factors, predicting survival and therapeutic responses, have been suggested. However, their consistency has not been demonstrated across studies. Considering the various underlying conditions of non-selected patients, further retrospective studies, that accurately reflect real-world situations, are needed to identify clinical biomarkers, that directly predict the favorable outcomes of ibrutinib treatment, and treatment strategies need to be established accordingly.

The optimal timing of ibrutinib treatment is unknown due to the lack of consensus and the introduction of various novel agents. Owing to the limited efficacy of ibrutinib in achieving and maintaining disease remission, most patients experience disease progression, leading to poor survival. Chimeric Antigen Receptor (CAR)-T cell therapy is recommended for these patients [16, 17]. However, real-world constraints limit the accessibility of this cutting-edge treatment to a subset of patients. Thus, it is challenging to find definitive and palliative treatment schemes for patients with progressive disease due to the changing trends in the treatment of MCL treatment, caused by the development of various targeted agents and immune-cellular therapies.

Therefore, this retrospective study evaluated the clinical outcomes of 42 patients with RR-MCL, who received ibrutinib. This study presents the outcomes of ibrutinib treatment according to associated clinical variables and discusses subsequent therapeutic strategies for patients, who were refractory to ibrutinib treatment. In addition, the overall response and survival outcomes of patients, following the continuation and discontinuation of ibrutinib, were investigated.

Study overview

This retrospective study investigated the characteristics and survival outcomes of patients with RR-MCL, treated with ibrutinib after at least one prior systemic therapy. By analyzing this comprehensive dataset, this study aimed to determine the benefits and outcomes, associated with salvage ibrutinib therapy in patients with RR-MCL. The study population was drawn from two prospective cohort studies (Institutional Review Board numbers: 2016-11-040 and 2022-05-078). Following a meticulous review of all tissue samples by pathology experts at our institution (J.C. and Y.H.K.), the diagnosis of MCL was conclusively established according to the WHO 2016 classification [18].

Patients and data collection

The medical records of the patients were reviewed to collect the following clinical and laboratory data: age; sex; Eastern Cooperative Oncology Group Performance Status; Ann Arbor stage; presence of extra-nodal involvement; bone marrow (BM) involvement; splenomegaly, defined by a horizontal length greater than 11 cm; Ki-67 expression rate; sMIPI; complete blood count; LDH; and β-2 microglobulin (B2M). Unfortunately, TP53 mutation evaluation using next- generation sequencing (NGS) was not performed in all patients due to lack of consent.

The study registration of patients was concluded in December 2021, and the data analysis was based on information available up to April 2023, which served as the cut-off date for this study. The IRB of the Samsung Medical Center approved this study (approval number. 2016-11- 040-025), and all participants provided written informed consent before their enrollment. The study adhered to the ethical principles, outlined in the Declaration of Helsinki and the Korea Good Clinical Practice guidelines. Ethical considerations were strictly followed throughout this study.

Ibrutinib therapy and response evaluation

Patients with RR-MCL received a prescribed daily dose of 560 mg of oral ibrutinib until disease progression or the emergence of intolerable toxicity. Dose adjustments, delays, and treatment discontinuation were decided at the physician’s discretion, based on the severity of adverse events (AEs) or disease progression. According to the Lugano classification, treatment response was evaluated using computed tomography (CT) and/or 18F-fluorodeoxyglucose positron- emission tomography CT [19]. Patients, who achieved a CR or partial response (PR), were categorized as responders, while those with stable disease (SD) or disease progression were considered non-responders. The patients, who did not achieve disease control (CR, PR, or SD), were classified as primary non-responders. Moreover, those, with controlled diseases, that receded over time, were classified as secondary non-responders. Finally, those, whose controlled disease status was maintained until the last follow-up, were classified as consistent responders. Early relapse was defined as disease recurrence within 12 months of ibrutinib administration.

The administration of the following first-line treatment options was confirmed: bendamustine and rituximab (BR); rituximab, cyclophosphamide, vincristine, doxorubicin, and prednisolone (R-CHOP); and rituximab, cyclophosphamide, cytarabine, vincristine, and prednisolone (R-Hyper-CVAD). The regimens were classified as BR- or anthracycline-containing (R-CHOP or R-Hyper-CVAD) for analysis. None of the patients received cytarabine-containing first-line chemo-therapy. A few patients eligible for autologous stem cell transplantation (ASCT) underwent high-dose chemotherapy, including cytarabine, followed by ASCT. None of the patients underwent allogeneic SCT.

Statistical analysis

Descriptive statistics, including proportions and medians, were used to analyze the data. Intergroup comparisons of categorical variables were performed using the Fisher’s exact test. ORR was defined as the proportion of patients, who achieved CR or PR, with the best response obtained during ibrutinib treatment. PFS and OS were evaluated using the Kaplan-Meier method. The PFS was measured from the initiation of ibrutinib treatment to the date of disease progression or death. OS was measured from the date of ibrutinib initiation to the date of death due to any cause or last follow-up. Additionally, the OS of post-ibrutinib treatment patients was calculated from the starting date of next-line anti-cancer therapies after ibrutinib interruption to the date of death from any cause or last follow-up. The cumulative incidence of early relapse was estimated using the Kaplan- Meier method. The log-rank test was used for comparisons, and both univariate and multivariate analyses were performed to estimate the associations between the survival rate and variables, concerning the administration of salvage ibrutinib. Data were analyzed using the Statistical Package for Social Sciences software (version 24.0, IBM Corp., Armonk, NY, USA).

Patient characteristics at diagnosis and outcomes of front-line treatment

The baseline demographics of the 42 patients at the time of their diagnosis are presented in Table 1. The median age was 62 years (range, 38–78 yr), and 25 patients (59.5%) were more than 60 years old. Most patients were male (N=30, 71.4%). Leukocytosis and elevated LDH levels were observed in 20% (N=11) and 45.4% (N=19) of the patients, respectively. Based on the pathologic reviews, most cases were documented as the classic MCL type (N=41, 97.6%), and 21.4% (N=9) presented with Ki-67 overexpression (≥30%). Among 37 patients (88.1%) diagnosed with stage III or IV disease, five (11.9%) presented with high-risk sMIPI, and most patients (N=37, 88.1%) were classified under low/intermediate- risk sMIPI.

Table 1 Baseline demographics and disease characteristics at diagnosis.

CharacteristicsTotal
(N=42)
BR
(N=16)
Anthracycline-based regimen (N=26)P
Age (yr)Median (range)62 (38–78)63 (50–78)61 (39–70)
<6017 (40.5%)2 (12.5%)15 (57.7%)0.004
≥6025 (59.5%)14 (87.5%)11 (42.3%)
GenderMale30 (71.4%)11 (68.7%)19 (73.0%)0.763
Female12 (28.6%)5 (31.2%)7 (27.0%)
ECOG-PS0–139 (92.9%)14 (87.5%)25 (96.2%)0.290
2–43 (7.1%)2 (12.5%)1 (3.8%)
B symptomAbsence37 (88.1%)13 (81.2%)24 (92.3%)0.283
Present5 (11.9%)3 (18.7%)2 (7.7%)
LeukocytosisAbsence31 (73.8%)8 (50.0%)23 (88.5%)0.006
Present11 (26.2%)8 (50.0%)3 (11.5%)
AnemiaAbsence33 (78.6%)9 (56.2%)24 (92.3%)0.006
Present9 (21.4%)7 (43.7%)2 (7.7%)
ThrombocytopeniaAbsence36 (85.7%)11 (68.7%)25 (96.2%)0.014
Present6 (14.3%)5 (31.2%)1 (3.8%)
Serum LDHAbsence23 (54.8%)7 (43.7%)16 (61.5%)0.261
Elevated19 (45.2%)9 (56.2%)10 (38.5%)
Serum β-2 microglobulinAbsence23 (54.8%)5 (31.2%)18 (69.2%)0.016
Elevated19 (45.2%)11 (68.7%)8 (30.8%)
Bone marrow involvementAbsence13 (31.0%)2 (12.5%)11 (42.3%)0.042
Present29 (69.0%)14 (87.5%)15 (57.7%)
Number of extra-nodal involvementLess than one11 (26.2%)2 (12.5%)9 (34.6%)0.113
More than two31 (73.8%)14 (87.5%)17 (65.4%)
Bulky (≥7 cm) massAbsence33 (78.6%)10 (62.5%)23 (88.5%)0.046
Present9 (21.4%)6 (37.5%)3 (11.5%)
PathologyClassic type41 (97.6%)16 (100.0%)25 (96.2%)0.427
Pleomorphic/blastoid type1 (2.4%)0 (0.0%)1 (3.8%)
Ki-67<30%33 (78.6%)12 (75.0%)21 (80.8%)0.658
≥30%9 (21.4%)4 (25.0%)5 (19.2%)
Ann Arbor stageI/II5 (11.9%)1 (6.3%)4 (15.4%)0.375
III/IV37 (88.1%)15 (93.7%)22 (84.6%)
sMIPI scoreLow (0–3)19 (45.2%)3 (18.7%)16 (61.5%)0.002
Intermediate (4–5)18 (42.9%)8 (50.0%)10 (38.5%)
High (6–11)5 (11.9%)5 (31.3%)0 (0.0%)

Abbreviations: ASCT, autologous stem cell transplantation; BR, bendamustine plus rituximab; ECOG-PS, Eastern Cooperative Oncology Group Performance Status; LDH, lactate dehydrogenase; sMIPI, simplified mantle cell lymphoma international prognostic index.



Among the 42 patients, 16 (38.1%) received BR, and 26 (61.9%) were treated with an anthracycline-containing regimen as a front-line treatment. Patients older than 60 years (P=0.004) with leukocytosis (P=0.006), anemia (P=0.006), thrombocytopenia (P=0.014), elevated B2M (P=0.016), BM involvement (P=0.042), or low-/intermediate-risk sMIPI (P=0.002) mainly received BR regimens rather than anthracycline-containing regimens (Table 1).

Ibrutinib as a salvage treatment

The patient’s characteristics at the start of ibrutinib treatment are presented in Supplementary Table 1. The patients received a median of one other treatment (range, 1–5) before ibrutinib administration. The ORR was 69% (N=29), with 21 CR (50%) and eight PR (19%). Patients (N=29, 69.0%), who had previously received one line of treatment, were administered a median of 12 cycles of ibrutinib (range, 1–55), and they achieved an ORR of 72.4% (N=21), with 16 CR (55.1%) and five PR (17.2%). Furthermore, patients (N=13, 31.0%), previously treated with more than two lines of treatment, received a median of 14 cycles of ibrutinib (range, 3–23), and they achieved an ORR of 61.5% (N=8), with five CR (38.4%) and three PR (23%). The most common reasons for treatment discontinuation were disease progression (N=23, 54.8%) and severe AEs (N=5, 11.9%).

During a median follow-up of 46.0 months (95% CI, 1.9–80.5), the median PFS was 16.4 months (95% CI, 0.0–41.5), and the median OS was 50.1 months (95% CI, 9.7–90.5; Fig. 1A, B). The responders showed superior PFS (46.0 mo; 95% CI, 26.2–65.7 vs. 7.9 mo; 95% CI, 2.8–12.9; P=0.001) and OS (63.5 mo; 95% CI, 42.9–67.3 vs. 13.6 mo; 95% CI, 7.3–19.9; P=0.002), compared to poorly responding patients (Fig. 1C, D). There was no difference in PFS (P=0.730) or OS (P=0.544) according to the line of treatment (Fig. 1E, F). However, patients, who experienced disease progression within 24 months of front-line treatment (POD 24), exhibited inferior PFS (10.5 mo; 95% CI, 4.4–16.6 vs. 53.8 mo; 95% CI, 27.9–79.6; P=0.005) and OS (12.6 mo; 95% CI, 4.5–20.7 vs. NR; P=0.015) upon receiving salvage ibrutinib treatment (Fig. 1G, H).

Fig. 1. Kaplan-Meier curve of progression-free survival (PFS) (A) and overall survival (OS) for salvage ibrutinib therapy (B). Comparison of PFS and OS of ibrutinib treatment according to response (C, D), line of treatment (E, F), and progression of disease within 24 months of front-line treatment (G, H). Assessment of PFS according to age (I), presence of splenomegaly (J), and simplified mantle cell lymphoma international prognostic index (sMIPI) (K) and OS according to age (L), presence of splenomegaly (M), and sMIPI (N).

Patients, who were older than 70 years at the initiation of ibrutinib treatment (7.9 mo; 95% CI, 1.3–14.4 vs. 36.6 mo; 95% CI, 9.8–63.5; P=0.004), or those with splenomegaly (9.9 mo; 95% CI, 0.4–19.4 vs. 46.0 mo; 95% CI, 10.3–81.6; P=0.022) or a high-risk sMIPI (5.0 mo; 95% CI, 2.3–7.7 vs. 37.4 mo; 95% CI, 29.4–45.4 vs. 63.5 mo; 95% CI, 0.0–152.8; P<0.001) exhibited inferior PFS with ibrutinib treatment (Fig. 1I–K). Moreover, patients older than 70 years (13.6 mo; 95% CI, 6.7–20.5 vs. 63.5 mo; 95% CI, 9.8–63.5; P=0.006) or those with splenomegaly (12.6 mo; 95% CI, 0.0–34.0 vs. not reached [NR]; P=0.006) or a high-risk sMIPI (9.9 mo; 95% CI, 3.4–16.4 vs. NR vs. NR; P<0.001) displayed poorer OS (Fig. 1L–N). In the univariate and multivariate analyses, the duration of ibrutinib treatment and sMIPI were significant factors, influencing both OS and PFS (Table 2).

Table 2 Univariate and multivariate analyses for progressiong-free survival and overall survival with salvage ibrutinib treatment.

VariablesOverall survivalProgression-free survival
UnivariateMultivariateUnivariateMultivariate
HR (95% CI)PHR (95% CI)PHR (95% CI)PHR (95% CI)P
Age (≥70 yr)3.419 (1.340–8.726)0.0100.380 (0.106–1.358)0.1373.158 (1.395–7.146)0.0060.578 (0.201–1.661)0.308
sMIPI score (high score)5.325 (2.111–13.428)<0.0016.249 (2.067–18.892)0.0014.681 (2.086–10.501)<0.0012.874 (1.171–7.055)0.021
Splenomegaly3.303 (1.345–8.111)0.0091.983 (0.439–8.961)0.3742.338 (1.107–4.936)0.0261.373 (0.506–3.720)0.534
Anthracycline-contai- ning front-line treatment0.197 (0.075–0.516)0.0011.282 (0.354–4.647)0.7050.159 (0.063–0.402)<0.0010.273 (0.081–0.921)0.036
POD 24 of front-line treatment3.013 (1.190–7.625)0.0200.991 (0.242–4.070)0.9912.858 (1.336–6.117)0.0071.895 (0.737–4.876)0.185
Previous number of treatments (≥2 lines )0.730 (0.263–2.024)0.5461.230 (0.576–2.626)0.593
Underwent ASCT before ibrutinib treatment0.692 (0.230–2.086)0.5131.068 (0.468–2.439)0.876
Duration of ibrutinib >12 months0.023 (0.003–0.179)<0.0010.011 (0.001–0.110)<0.0010.041 (0.011–0.154)<0.0010.042 (0.010–0.180)<0.001
Grade 3/4 adverse events3.185 (1.203–8.433)0.0201.884 (0.612–5.798)0.2691.977 (0.789–4.953)0.146

Abbreviations: ASCT, autologous stem cell transplantation; POD 24, disease progression within 24 months; sMIPI, simplified mantle cell lymphoma international prognostic index.



Characteristics of the patients who experienced early relapse

An analysis was conducted to confirm the clinical markers, affecting early relapse during ibrutinib treatment (N=13, 30.9%). According to the independent samples t-test, only a high-risk sMIPI was correlated with early relapse (P=0.033, Table 3). Patients with early relapse had significantly poorer survival outcomes than those, who did not experience early relapse (12.6 mo; 95% CI, 7.9–17.3 vs. NR; P<0.001, Fig. 2A). The primary non-responders had exceptionally poor survival compared to secondary non-responders and consistent responders (5.9 mo; 95% CI, 0.7–11.1 vs. 30.7 mo; 95% CI, 1.2–60.2 vs. NR; P=0.002, Fig. 2B). In terms of cumulative incidence, patients who received BR as front-line treatment (P=0.004), those with high-risk sMIPI (P=0.010), and those older than 70 years (P=0.011) were strongly correlated with an increased cumulative incidence of early relapse (Fig. 2C–E). However, there was a weak correlation between early relapse, splenomegaly (P=0.084), and prior lines of treatment (P=0.720) (Fig. 2F, G).

Fig. 2. Kaplan-Meier curves of overall survival according to early relapse (A) and type of response to salvage ibrutinib (B). Comparison of cumulative incidence of early relapse according to front-line treatment (C), simplified mantle cell lymphoma international prognostic index (D), age (E), presence of splenomegaly (F), and line of treat-ment of ibrutinib (G).

Table 3 Comparison of patients with and without early disease relapse.

CharacteristicsEarly relapse (N=13)Late relapse or no relapse (N=29)P
Age (yr)Median (yr), range67 (51–77)67 (45–81)
<706 (46.2%)21 (72.4%)0.101
≥707 (53.8%)8 (27.6%)
GenderMale10 (76.9%)20 (69.0%)0.598
Female3 (23.1%)9 (31.0%)
ECOG-PS0–112 (92.3%)29 (100.0%)0.131
2–41 (7.7%)0 (0.0%)
sMIPI scoreLow (0–3)0 (0.0%)7 (24.1%)0.033
Intermediate (4–5)6 (46.2%)14 (48.3%)
High (6–11)7 (53.8%)8 (27.6%)
Splenomegaly (≥11 cm)Absence7 (53.8%)11 (37.9%)0.335
Present6 (46.2%)18 (62.1%)
Prior number of treatmentPrior 1 line9 (69.2%)20 (69.0%)0.986
Prior ≥2 lines4 (30.8%)9 (31.0%)
Front-line treatmentAnthracycline-based6 (46.2%)20 (69.0%)0.159
Bendamustine-based7 (53.8%)9 (31.0%)
ASCTReceived3 (23.1%)7 (24.1%)0.941
Not received10 (76.9%)22 (75.9%)
Ibrutinib dose adjustmentDose reduction1 (7.7%)5 (17.2%)0.414
No dose reduction12 (92.3%)24 (82.8%)
Ibrutinib discontinuationDiscontinuation13 (100.0%)24 (82.8%)0.111
No discontinuation0 (0.0%)5 (17.2%)

Abbreviations: ASCT, autologous stem cell transplantation; ECOG-PS, Eastern Cooperative Oncology Group Performance Status; sMIPI, simplified mantle cell lymphoma international prognostic index.



Post-ibrutinib survival outcomes

The median OS for patients, who discontinued ibrutinib treatment (N=37, 88.1%), was 9.8 months (95% CI, 0.6–19.0) (Fig. 3A). The ibrutinib responders had a better post-ibrutinib OS than the non-responders, but the difference was not statistically significant (20.0 mo; 95% CI, 0.0–43.0 vs. 5.7 mo; 95% CI, 0.0–13.2; P=0.101). The primary non-responders, who did not achieve disease control, had a median OS of only 2.6 months (95% CI, 0.0–10.4) after ibrutinib treatment.

Fig. 3. Kaplan-Meier curves of overall survival for post-ibrutinib treatment (A). Comparison of overall survival according to participation in clinical trials (B). Swimmer plot of patients who received post-ibrutinib treatment (C).

Twenty-three patients (62.1%) received additional anti-cancer therapies after ibrutinib discontinuation, and the estimated median OS after ibrutinib was 12.2 months (95% CI, 2.1–22.2). Sixty-five percent of patients (N=15) received bortezomib or novel agents, such as lenalidomide and venetoclax, and 21.7% (N=5) participated in clinical trials. Although statistical significance was not achieved, the patients assigned to clinical trials had improved survival outcomes, compared to those, who received conventional chemotherapy (P=0.458, Fig. 3B). In terms of post-ibrutinib survival, the patients, who experienced early relapse, generally had an inferior OS (5.4 mo; 95% CI, 0.0–12.7 vs. 24.8 mo; 95% CI, 15.7–37.2; P=0.012) (Fig. 3C).

Adverse events with ibrutinib treatment

During ibrutinib treatment, various hematologic and non-hematologic AEs were observed, but most were reported as grade 1 or 2 and were manageable. Six patients (14.3%) received a reduced ibrutinib dose, and five patients (11.9%) discontinued ibrutinib treatment due to toxicities (Supplementary Table 2). All grades of thrombocytopenia accounted for 11.9% of the hematologic AEs (N=5). In particular, grade ≥3 thrombocytopenia accounted for 7.1% (N=3). The incidence of all grades of anemia and neutropenia was 7.1%, and less than 5% had a grade ≥3. The most common non-hematologic AE was skin rash (N=20, 47.6%), and only four patients (9.5%) presented with a grade ≥3. Grade ≥3 fatigue and diarrhea were observed in approximately 9.5% (N=4) and 7.1% (N=3), respectively. Three patients (7.1%) had grade 1 or 2 atrial fibrillation, and all were adequately managed through medical intervention. Of the five patients who discontinued ibrutinib, one ceased treatment due to a hematologic AE (grade 3 thrombocytopenia), while the remaining four stopped due to non-hematologic AEs (two patients with grade 3 diarrhea, one with grade 3 rash, and one with grade 3 fatigue). Among the consistent responders older than 70 years, those who were maintained on ibrutinib treatment with well-managed severe AEs had a more favorable PFS (36.6 mo; 95% CI, 12.3–60.9 vs. 11.4 mo; 95% CI, 0.0–23.1; P=0.234) and OS (NR vs. 11.4 mo; 95% CI, 0.0–23.1; P=0.062) (Supplementary Fig. 1A).

This study evaluated the efficacy and survival outcomes of ibrutinib treatment, based on real-world clinical experience. According to previous studies (Table 4) [5-14],

Table 4 Summary of previous studies on ibrutinib therapy in relapsed or refractory mantle cell lymphoma.

StudyPt. numberFront-line Tx.Refractory to front-line Tx.Upfront ASCTPrior Tx.ORRPFS (median)OS (median)Ref
Phase 2111R-containing therapy (N=99, 89%)50 (45%)8 (13%)3 (1–5)67% (CR, 23%)13 mo22.5 mo[5, 6]
Phase 3280R-containing therapy (N=280, 100%)36 (26%)N/A2 (1–9)72% (CR, 19%)14.6 moNot reached[7]
Retrospective97Intensive induction therapy (N=52, 54%) (R±hyper-CVAD, R-maxi CHOP)7 (7%)38 (39%)2 (1–8)65% (CR, 33%)15 mo22 mo[8]
Phase 216R-containing therapy (N=16, 100%)1 (6.2%)N/A2.5 (1–4)87.5% (CR, 12.5%)N/AN/A[9]
Retrospective33R-CHOP (N=33)N/A6 (18.1%)

1 (33.3%)

2 (36.4%)

≥3 (30.3%)

64% (CR, 15%)27.4 mo35.1 mo[10]
Retrospective65R-CHOP, R-bendamustine and R-hyper-CVAD (87%)N/AN/A2 (1–6)N/A12.0 mo18.5 mo[11]
Retrospective88

R±CHOP (N=63)

R±hyper-CVAD (N=13)

BR (N=6)

VR-CAP (N=3)

CVP/bendamustine (N=3)

N/A5 (5.6%)1 (1–6)64.8% (CR, N/A)20.8 mo2-yr OS rate 79.5%[12]
Retrospective211

R-CHOP/mini-CHOP (N=70, 33%)

Cytarabine-based regimen (N=60, 28%)

R-bendamustine (N=45, 21%)

VR-CAP (N=5, 2%)

Others (N=31, 15%)

38 (18%)53 (25%)169% (CR, 27%)17.8 mo23.9 mo[13]
Retrospective77

CHOP/CHOP-like (N=42, 54%)

Cytarabine-based regimen (N=25, 33%)

Non-anthracycline regimen (N=10, 13%)

15 (20%)19 (25%)

1 (29%)

2 (30%)

≥3 (41%)

66%

(CR, 31%)

10.3 mo23.1 mo[14]
Retrospective42

R-CHOP (N=18, 43%)

R-hyper-CVAD (N=8, 19%)

R-bendamustine (N=16, 38%)

20 (47.6%)6 (14.3%)1 (1–5)69% (CR, 50%)19.2 mo50.1 moCurrent study

Abbreviations: ASCT, autologous stem cell transplantation; BR, rituximab, bendamustine; CHOP, cyclophosphamide, doxorubicin, vincristine, prednisolone; CVP, cyclophosphamide, vincristine, prednisone; Hyper-CVAD, hyperfractionated cyclophosphamide, vincristine, doxorubicin dexamethasone; LOT, line of therapy; N/A, non-available; ORR, objective response rate; OS, overall survival; PFS, progression-free survival; Pt., patient; R, rituximab; Tx., treatment; VR-CAP, bortezomib, rituximab, cyclophosphamide, doxorubicin, prednisolone.



Based on the CAN3001 study, which reported the long-term outcomes of salvage ibrutinib treatment with 10 years of follow-up [20], severe treatment-related AEs (TRAEs) were generally observed within one year of ibrutinib initiation, but it decreased annually thereafter. The AEs reported in this study were comparable to those, documented in previous studies, in terms of the type, timing of occurrence, and severity. This underscores that the prompt and proactive management of AEs during ibrutinib treatment alleviates the patient’s symptoms and enhances survival outcomes. The patients, who received active toxicity management along with ibrutinib, had a stable treatment course and improved clinical outcomes despite AEs. Thus, to prevent treatment interruption and achieve favorable outcomes, acute and chronic AEs, associated with ibrutinib therapy, should be recognized, monitored, and managed.

In this study, ibrutinib monotherapy resulted in an ORR of approximately 70%, which was similar to or higher than the response rates, reported in other studies (Table 4). Ibrutinib was more efficacious in patients, older than 70 years, who received anthracycline-containing front-line treatment, than in those, who received bendamustin-based front-line treatment (Supplementary Fig. 1B). Patients, treated with a bendamustine-containing regimen, presented with unfavorable baseline characteristics, including age >70 years, elevated WBC count, BM involvement, and high-risk sMIPI at the time of diagnosis (Table 1). Based on this, less intensive chemotherapy is preferred as the front-line treatment for patients aged 70 or older as they have poor general health conditions or comorbidities. It was assumed that prior bendamustine exposure led to poor subsequent ibrutinib treatment outcomes, as observed in other studies [10]. In cases where POD 24 occurred, regardless of the front-line treatment, favorable survival outcomes were not guaranteed, even with ibrutinib treatment. In line with previous studies [20, 21], POD 24 persisted as an unfavorable prognostic indicator in the targeted agent era.

TP53 mutation is a well-known biomarker, that predicts poor outcomes with conventional treatments in both newly diagnosed and RR-MCL patients. According to several retrospective studies [15, 22], the incidence of TP53 mutations was approximately 14%, and patients with TP53 mutations had lower response rates (55% vs. 70%) as well as shorter PFS (4 mo vs. 12 mo) and OS (10 mo vs. 34 mo) than patients with wild-type TP53. Unfortunately, this study was unable to include data on TP53 mutation status due to limited tissue samples and the lack of patient consent. While the presence of TP53 mutations impacts the prognosis of MCL, performing NGS on all MCL patients was not cost-effective in a real-world setting since only 14% were found to have TP53 mutations. Instead, Eskelund et al. [23] suggested specific clinical factors, related to TP53 mutation. These include a blastoid morphology, high-risk MIPI, and Ki67 >30%. In cases, where determining the TP53 mutation status is challenging, establishing a treatment plan, based on the implied factors, and defining the clinical signs of TP53 mutations are essential.

In this study, early disease relapse was defined as disease relapse within 12 months of starting ibrutinib treatment since the reported median PFS of ibrutinib was 12.6 months (95% CI, 10.3–16.6). Patients with early relapse exhibited significantly shorter PFS than those, who did not experience early relapse (5.4 mo vs. 24.8 mo). High-risk sMIPI scores were correlated with early disease relapse. Unfortunately, these patients were unsuitable for any subsequent treatment due to rapid disease deterioration, and the post-ibrutinib survival outcomes were poor. Establishing optimal treatment strategies for patients, who were refractory to ibrutinib, remains challenging. Allogeneic SCT has demonstrated long-term disease control through the graft-to-host effect, but it is limited to patients with favorable general conditions for withstanding intensive induction therapy and life-threatening complications. Pirtobrutinib, a novel non-covalent BKT inhibitor, achieved an ORR of 58%. Patients with RR-MCL, who received a median of three prior treatment modalities, including ibrutinib, responded for 21.6 months [24]. Moreover, brexucabtagene autoleucel (brex-cel), an anti-CD19 CAR-T cell therapy, achieved an ORR of 93% and CR rate of 67% [16, 17]. After 12 months, 57% of patients remained in remission. The lisocaltagene maraleucel (liso-cel) was associated with a low incidence of grade ≥3 TRAEs, and it demonstrated promising clinical activity in a phase 1 study [25]. Thus, novel therapeutic approaches, including targeted agents and immunocellular therapies, should be utilized in the management of patients, experiencing early disease relapse. Additionally, the risk stratification system should be continuously validated and adjusted to improve treatment outcomes.

In conclusion, this study demonstrated the efficacy and safety of salvage ibrutinib in patients with RR-MCL, and the patients, who potentially benefit from salvage ibrutinib therapy, were identified by verifying prognostic factors. Favorable survival outcomes were more frequent among patients with an objective response or those, who remained in disease remission with continuous ibrutinib treatment. To maximize the benefits of ibrutinib treatment, the capacity of the patient to endure TRAEs and the specific characteristics of their disease should be considered. Since patients, who experience early disease relapse within 12 months, have poorer survival outcomes, groundbreaking treatment strategies should be identified.

No potential conflicts of interest relevant to this article were reported.

  1. Armitage JO, Longo DL. Mantle-cell lymphoma. N Engl J Med 2022;386:2495-506.
    Pubmed CrossRef
  2. Lee H, Park HJ, Park EH, et al. Nationwide statistical analysis of lymphoid malignancies in Korea. Cancer Res Treat 2018;50:222-38.
    Pubmed KoreaMed CrossRef
  3. Merolle MI, Ahmed M, Nomie K, Wang ML. The B cell receptor signaling pathway in mantle cell lymphoma. Oncotarget 2018;9:25332-41.
    Pubmed KoreaMed CrossRef
  4. Gu D, Tang H, Wu J, Li J, Miao Y. Targeting Bruton tyrosine kinase using noncovalent inhibitors in B cell malignancies. J Hematol Oncol 2021;14:40.
    Pubmed KoreaMed CrossRef
  5. Wang ML, Rule S, Martin P, et al. Targeting BTK with ibrutinib in relapsed or refractory mantle-cell lymphoma. N Engl J Med 2013;369:507-16.
    Pubmed KoreaMed CrossRef
  6. Wang ML, Blum KA, Martin P, et al. Long-term follow-up of MCL patients treated with single-agent ibrutinib: updated safety and efficacy results. Blood 2015;126:739-45.
    Pubmed KoreaMed CrossRef
  7. Dreyling M, Jurczak W, Jerkeman M, et al. Ibrutinib versus temsirolimus in patients with relapsed or refractory mantle-cell lymphoma: an international, randomised, open-label, phase 3 study. Lancet 2016;387:770-8.
    Pubmed CrossRef
  8. Epperla N, Hamadani M, Cashen AF, et al. Predictive factors and outcomes for ibrutinib therapy in relapsed/refractory mantle cell lymphoma-a "real world" study. Hematol Oncol 2017;35:528-35.
    Pubmed CrossRef
  9. Maruyama D, Nagai H, Fukuhara N, et al. Efficacy and safety of ibrutinib in Japanese patients with relapsed or refractory mantle cell lymphoma. Cancer Sci 2016;107:1785-90.
    Pubmed KoreaMed CrossRef
  10. Jeon YW, Yoon S, Min GJ, et al. Clinical outcomes for ibrutinib in relapsed or refractory mantle cell lymphoma in real-world experience. Cancer Med 2019;8:6860-70.
    Pubmed KoreaMed CrossRef
  11. Tucker D, Morley N, MacLean P, et al. The 5-year follow-up of a real-world observational study of patients in the United Kingdom and Ireland receiving ibrutinib for relapsed/refractory mantle cell lymphoma. Br J Haematol 2021;192:1035-8.
    Pubmed CrossRef
  12. Yi JH, Kim SJ, Yoon DH, et al. Real-world outcomes of ibrutinib therapy in Korean patients with relapsed or refractory mantle cell lymphoma: a multicenter, retrospective analysis. Cancer Commun (Lond) 2021;41:275-8.
    Pubmed KoreaMed CrossRef
  13. McCulloch R, Lewis D, Crosbie N, et al. Ibrutinib for mantle cell lymphoma at first relapse: a United Kingdom real-world analysis of outcomes in 211 patients. Br J Haematol 2021;193:290-8.
    Pubmed CrossRef
  14. Obr A, Benesova K, Janikova A, et al. Ibrutinib in mantle cell lymphoma: a real-world retrospective multi-center analysis of 77 patients treated in the Czech Republic. Ann Hematol 2023;102:107-15.
    Pubmed KoreaMed CrossRef
  15. Rule S, Dreyling M, Goy A, et al. Ibrutinib for the treatment of relapsed/refractory mantle cell lymphoma: extended 3.5-year follow up from a pooled analysis. Haematologica 2019;104:e211-4.
    Pubmed KoreaMed CrossRef
  16. Wang M, Munoz J, Goy A, et al. KTE-X19 CAR T-cell therapy in relapsed or refractory mantle-cell lymphoma. N Engl J Med 2020;382:1331-42.
    Pubmed KoreaMed CrossRef
  17. Wang M, Munoz J, Goy A, et al. Three-year follow-up of KTE-X19 in patients with relapsed/refractory mantle cell lymphoma, including high-risk subgroups, in the ZUMA-2 study. J Clin Oncol 2023;41:555-67.
    Pubmed KoreaMed CrossRef
  18. Swerdlow SH, Campo E, Pileri SA, et al. The 2016 revision of the World Health Organization classification of lymphoid neoplasms. Blood 2016;127:2375-90.
    Pubmed KoreaMed CrossRef
  19. Cheson BD, Fisher RI, Barrington SF, et al. Recommendations for initial evaluation, staging, and response assessment of Hodgkin and non-Hodgkin lymphoma: the Lugano classification. J Clin Oncol 2014;32:3059-68.
    Pubmed KoreaMed CrossRef
  20. Dreyling M, Goy A, Hess G, et al. Long-term outcomes with ibrutinib treatment for patients with relapsed/refractory mantle cell lymphoma: a pooled analysis of 3 clinical trials with nearly 10 years of follow-up. Hemasphere 2022;6:e712.
    Pubmed KoreaMed CrossRef
  21. Bond DA, Switchenko JM, Maddocks KJ, et al. Outcomes following early relapse in patients with mantle cell lymphoma. Blood (ASH Annual Meeting Abstracts) 2019;134(Suppl 1):753.
    CrossRef
  22. Lew TE, Minson A, Dickinson M, et al. Treatment approaches for patients with TP53-mutated mantle cell lymphoma. Lancet Haematol 2023;10:e142-54.
    Pubmed CrossRef
  23. Eskelund CW, Dahl C, Hansen JW, et al. TP53 mutations identify younger mantle cell lymphoma patients who do not benefit from intensive chemoimmunotherapy. Blood 2017;130:1903-10.
    Pubmed CrossRef
  24. Wang ML, Jurczak W, Zinzani PL, et al. Pirtobrutinib in covalent bruton tyrosine kinase inhibitor pretreated mantle-cell lymphoma. J Clin Oncol 2023;41:3988-97.
    Pubmed KoreaMed CrossRef
  25. Palomba ML, Gordon LI, Siddiqi T, et al. Safety and preliminary efficacy in patients with relapsed/refractory mantle cell lymphoma receiving lisocabtagene maraleucel in transcend NHL 001. Blood (ASH Annual Meeting Abstracts) 2020;136(Suppl 1):10-1.
    CrossRef

Article

Original Article

Blood Res 2023; 58(4): 208-220

Published online December 31, 2023 https://doi.org/10.5045/br.2023.2023208

Copyright © The Korean Society of Hematology.

A retrospective analysis of ibrutinib outcomes in relapsed or refractory mantle cell lymphoma

Yong-Pyo Lee1#, Ye Ji Jung2#, Junhun Cho3, Young Hyeh Ko3, Won Seog Kim2,4, Seok Jin Kim2,4, Sang Eun Yoon2

1Division of Hematology-Oncology, Department of Internal Medicine, Chungbuk National University Hospital, Cheongju, Division of Hematology-Oncology, Departments of 2Medicine, 3Pathology, Samsung Medical Center, Sungkyunkwan University School of Medicine, 4Health Sciences and Technology, Samsung Advanced Institute for Health Sciences and Technology, Sungkyunkwan University, Seoul, Korea

Correspondence to:Sang Eun Yoon, M.D., Ph.D.
Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, 81 Irwon-ro, Gangnam-gu, Seoul 06351, Korea
E-mail: starload0326@gmail.com

#These authors contributed equally to this work.

*This study was supported by a grant from the Korea Health Technology R&D Project through the Korea Health Industry Development Institute (KHIDI), funded by the Ministry of Health and Welfare, Republic of Korea (grant number: HI22C0999). This study was also supported by grants from the Korea Health Technology R&D Project through the Korea Health Industry Development Institute, funded by the Ministry of Health & Welfare, Republic of Korea (HR20C0025); a National Research Foundation of Korea grant funded by the Korean government (2022R1F1A1064058); and the Bio&Medical Technology Development Program of the National Research Foundation (NRF) funded by the Korean government (MSIT) (No. RS-2023-00222838).

Received: October 26, 2023; Revised: November 27, 2023; Accepted: December 8, 2023

This is an Open Access article distributed under the terms of the Creative Commons Attribution Non-Commercial License (http://creativecommons.org/licenses/by-nc/4.0) which permits unrestricted non-commercial use, distribution, and reproduction in any medium, provided the original work is properly cited.

Abstract

Background
While treatment strategies for mantle cell lymphoma (MCL) have evolved, patients often experience disease progression and require additional treatment therapies. Ibrutinib presents a promising option for relapsed or refractory MCL (RR-MCL). This study investigated real-world treatment outcomes of ibrutinib in patients with RR-MCL.
Methods
A single-center retrospective analysis investigated clinical characteristics and survival outcomes of patients with RR-MCL, treated with ibrutinib.
Results
Forty-two patients were included, with 16 received rituximab and bendamustine, and 26 receiving anthracycline-based regimens as front-line treatment. During a median follow-up of 46.0 months, the response rate to ibrutinib was 69%, with 12 CRs and 8 partial responses. Disease progression (54.8%) and adverse events (11.9%) were the primary reasons for discontinuation. Median progression-free survival (PFS) and overall survival (OS) were approximately 16.4 and 50.1 months, respectively. Patients older than 70 years (P=0.044 and P=0.006), those with splenomegaly (P=0.022 and P=0.006), and those with a high-risk simplified Mantle Cell Lymphoma International Prognostic Index (sMIPI) (P<0.001 and P<0.001) exhibited siginificantly inferior PFS and OS. Notably, patients with a high-risk sMIPI relapsed earlier. Post-ibrutinib treatment yilded an OS of 12.2 months, while clinical trial participants demonstrated superior survival compared to those receiving chemotherapy alone.
Conclusion
This study underscores the importance of considering patient characteristics before administering ibrutinib as salvage therapy. Early relapse was associated with poor outcomes, highlighting the need for novel therapeutic strategies.

Keywords: Ibrutinib, Mantle-cell, Lymphoma, Relapsed or refractory

INTRODUCTION

Mantle cell lymphoma (MCL) is a distinct subtype of non-Hodgkin’s, accounting for approximately 2% of B-cell malignancies [1, 2]. According to an understanding of the developmental etiology of MCL, deregulation of B-cell receptor (BCR) signaling and Bruton tyrosine kinase (BTK) permits continuous B-cell proliferation, leading to MCL development in the pre-germinal center. BTK inhibitors block BCR signaling propagation, related to B-cell proliferation, and allow cancer cell apoptosis by ultimately reducing nuclear factor-κB pathway activity [3]. Thus, BTK is a key factor in BCR signaling and a therapeutic target for MCL. Several covalent (ibrutinib, acalabrutinib, and zanubrutinib) and non-covalent (pirtobrutinib) BTK inhibitors have been developed [4].

Among the various BTK inhibitors, ibrutinib, the first oral selective BTK inhibitor, has a favorable objective response rate (ORR) of 65–70% with a complete response (CR) rate of 19% and progression-free survival (PFS) of 12–15 months for relapsed/refractory MCL (RR-MCL) patients, who received more than two treatments prior [5-15]. Previous studies suggested that the response to ibrutinib depended on the simplified Mantle Cell Lymphoma Inter-national Prognostic Index (sMIPI), which is calculated based on age, performance status, lactate dehydrogenase (LDH), and white blood cell (WBC) count. In addition to the sMIPI, other factors, predicting survival and therapeutic responses, have been suggested. However, their consistency has not been demonstrated across studies. Considering the various underlying conditions of non-selected patients, further retrospective studies, that accurately reflect real-world situations, are needed to identify clinical biomarkers, that directly predict the favorable outcomes of ibrutinib treatment, and treatment strategies need to be established accordingly.

The optimal timing of ibrutinib treatment is unknown due to the lack of consensus and the introduction of various novel agents. Owing to the limited efficacy of ibrutinib in achieving and maintaining disease remission, most patients experience disease progression, leading to poor survival. Chimeric Antigen Receptor (CAR)-T cell therapy is recommended for these patients [16, 17]. However, real-world constraints limit the accessibility of this cutting-edge treatment to a subset of patients. Thus, it is challenging to find definitive and palliative treatment schemes for patients with progressive disease due to the changing trends in the treatment of MCL treatment, caused by the development of various targeted agents and immune-cellular therapies.

Therefore, this retrospective study evaluated the clinical outcomes of 42 patients with RR-MCL, who received ibrutinib. This study presents the outcomes of ibrutinib treatment according to associated clinical variables and discusses subsequent therapeutic strategies for patients, who were refractory to ibrutinib treatment. In addition, the overall response and survival outcomes of patients, following the continuation and discontinuation of ibrutinib, were investigated.

MATERIALS AND METHODS

Study overview

This retrospective study investigated the characteristics and survival outcomes of patients with RR-MCL, treated with ibrutinib after at least one prior systemic therapy. By analyzing this comprehensive dataset, this study aimed to determine the benefits and outcomes, associated with salvage ibrutinib therapy in patients with RR-MCL. The study population was drawn from two prospective cohort studies (Institutional Review Board numbers: 2016-11-040 and 2022-05-078). Following a meticulous review of all tissue samples by pathology experts at our institution (J.C. and Y.H.K.), the diagnosis of MCL was conclusively established according to the WHO 2016 classification [18].

Patients and data collection

The medical records of the patients were reviewed to collect the following clinical and laboratory data: age; sex; Eastern Cooperative Oncology Group Performance Status; Ann Arbor stage; presence of extra-nodal involvement; bone marrow (BM) involvement; splenomegaly, defined by a horizontal length greater than 11 cm; Ki-67 expression rate; sMIPI; complete blood count; LDH; and β-2 microglobulin (B2M). Unfortunately, TP53 mutation evaluation using next- generation sequencing (NGS) was not performed in all patients due to lack of consent.

The study registration of patients was concluded in December 2021, and the data analysis was based on information available up to April 2023, which served as the cut-off date for this study. The IRB of the Samsung Medical Center approved this study (approval number. 2016-11- 040-025), and all participants provided written informed consent before their enrollment. The study adhered to the ethical principles, outlined in the Declaration of Helsinki and the Korea Good Clinical Practice guidelines. Ethical considerations were strictly followed throughout this study.

Ibrutinib therapy and response evaluation

Patients with RR-MCL received a prescribed daily dose of 560 mg of oral ibrutinib until disease progression or the emergence of intolerable toxicity. Dose adjustments, delays, and treatment discontinuation were decided at the physician’s discretion, based on the severity of adverse events (AEs) or disease progression. According to the Lugano classification, treatment response was evaluated using computed tomography (CT) and/or 18F-fluorodeoxyglucose positron- emission tomography CT [19]. Patients, who achieved a CR or partial response (PR), were categorized as responders, while those with stable disease (SD) or disease progression were considered non-responders. The patients, who did not achieve disease control (CR, PR, or SD), were classified as primary non-responders. Moreover, those, with controlled diseases, that receded over time, were classified as secondary non-responders. Finally, those, whose controlled disease status was maintained until the last follow-up, were classified as consistent responders. Early relapse was defined as disease recurrence within 12 months of ibrutinib administration.

The administration of the following first-line treatment options was confirmed: bendamustine and rituximab (BR); rituximab, cyclophosphamide, vincristine, doxorubicin, and prednisolone (R-CHOP); and rituximab, cyclophosphamide, cytarabine, vincristine, and prednisolone (R-Hyper-CVAD). The regimens were classified as BR- or anthracycline-containing (R-CHOP or R-Hyper-CVAD) for analysis. None of the patients received cytarabine-containing first-line chemo-therapy. A few patients eligible for autologous stem cell transplantation (ASCT) underwent high-dose chemotherapy, including cytarabine, followed by ASCT. None of the patients underwent allogeneic SCT.

Statistical analysis

Descriptive statistics, including proportions and medians, were used to analyze the data. Intergroup comparisons of categorical variables were performed using the Fisher’s exact test. ORR was defined as the proportion of patients, who achieved CR or PR, with the best response obtained during ibrutinib treatment. PFS and OS were evaluated using the Kaplan-Meier method. The PFS was measured from the initiation of ibrutinib treatment to the date of disease progression or death. OS was measured from the date of ibrutinib initiation to the date of death due to any cause or last follow-up. Additionally, the OS of post-ibrutinib treatment patients was calculated from the starting date of next-line anti-cancer therapies after ibrutinib interruption to the date of death from any cause or last follow-up. The cumulative incidence of early relapse was estimated using the Kaplan- Meier method. The log-rank test was used for comparisons, and both univariate and multivariate analyses were performed to estimate the associations between the survival rate and variables, concerning the administration of salvage ibrutinib. Data were analyzed using the Statistical Package for Social Sciences software (version 24.0, IBM Corp., Armonk, NY, USA).

RESULTS

Patient characteristics at diagnosis and outcomes of front-line treatment

The baseline demographics of the 42 patients at the time of their diagnosis are presented in Table 1. The median age was 62 years (range, 38–78 yr), and 25 patients (59.5%) were more than 60 years old. Most patients were male (N=30, 71.4%). Leukocytosis and elevated LDH levels were observed in 20% (N=11) and 45.4% (N=19) of the patients, respectively. Based on the pathologic reviews, most cases were documented as the classic MCL type (N=41, 97.6%), and 21.4% (N=9) presented with Ki-67 overexpression (≥30%). Among 37 patients (88.1%) diagnosed with stage III or IV disease, five (11.9%) presented with high-risk sMIPI, and most patients (N=37, 88.1%) were classified under low/intermediate- risk sMIPI.

Table 1 . Baseline demographics and disease characteristics at diagnosis..

CharacteristicsTotal
(N=42)
BR
(N=16)
Anthracycline-based regimen (N=26)P
Age (yr)Median (range)62 (38–78)63 (50–78)61 (39–70)
<6017 (40.5%)2 (12.5%)15 (57.7%)0.004
≥6025 (59.5%)14 (87.5%)11 (42.3%)
GenderMale30 (71.4%)11 (68.7%)19 (73.0%)0.763
Female12 (28.6%)5 (31.2%)7 (27.0%)
ECOG-PS0–139 (92.9%)14 (87.5%)25 (96.2%)0.290
2–43 (7.1%)2 (12.5%)1 (3.8%)
B symptomAbsence37 (88.1%)13 (81.2%)24 (92.3%)0.283
Present5 (11.9%)3 (18.7%)2 (7.7%)
LeukocytosisAbsence31 (73.8%)8 (50.0%)23 (88.5%)0.006
Present11 (26.2%)8 (50.0%)3 (11.5%)
AnemiaAbsence33 (78.6%)9 (56.2%)24 (92.3%)0.006
Present9 (21.4%)7 (43.7%)2 (7.7%)
ThrombocytopeniaAbsence36 (85.7%)11 (68.7%)25 (96.2%)0.014
Present6 (14.3%)5 (31.2%)1 (3.8%)
Serum LDHAbsence23 (54.8%)7 (43.7%)16 (61.5%)0.261
Elevated19 (45.2%)9 (56.2%)10 (38.5%)
Serum β-2 microglobulinAbsence23 (54.8%)5 (31.2%)18 (69.2%)0.016
Elevated19 (45.2%)11 (68.7%)8 (30.8%)
Bone marrow involvementAbsence13 (31.0%)2 (12.5%)11 (42.3%)0.042
Present29 (69.0%)14 (87.5%)15 (57.7%)
Number of extra-nodal involvementLess than one11 (26.2%)2 (12.5%)9 (34.6%)0.113
More than two31 (73.8%)14 (87.5%)17 (65.4%)
Bulky (≥7 cm) massAbsence33 (78.6%)10 (62.5%)23 (88.5%)0.046
Present9 (21.4%)6 (37.5%)3 (11.5%)
PathologyClassic type41 (97.6%)16 (100.0%)25 (96.2%)0.427
Pleomorphic/blastoid type1 (2.4%)0 (0.0%)1 (3.8%)
Ki-67<30%33 (78.6%)12 (75.0%)21 (80.8%)0.658
≥30%9 (21.4%)4 (25.0%)5 (19.2%)
Ann Arbor stageI/II5 (11.9%)1 (6.3%)4 (15.4%)0.375
III/IV37 (88.1%)15 (93.7%)22 (84.6%)
sMIPI scoreLow (0–3)19 (45.2%)3 (18.7%)16 (61.5%)0.002
Intermediate (4–5)18 (42.9%)8 (50.0%)10 (38.5%)
High (6–11)5 (11.9%)5 (31.3%)0 (0.0%)

Abbreviations: ASCT, autologous stem cell transplantation; BR, bendamustine plus rituximab; ECOG-PS, Eastern Cooperative Oncology Group Performance Status; LDH, lactate dehydrogenase; sMIPI, simplified mantle cell lymphoma international prognostic index..



Among the 42 patients, 16 (38.1%) received BR, and 26 (61.9%) were treated with an anthracycline-containing regimen as a front-line treatment. Patients older than 60 years (P=0.004) with leukocytosis (P=0.006), anemia (P=0.006), thrombocytopenia (P=0.014), elevated B2M (P=0.016), BM involvement (P=0.042), or low-/intermediate-risk sMIPI (P=0.002) mainly received BR regimens rather than anthracycline-containing regimens (Table 1).

Ibrutinib as a salvage treatment

The patient’s characteristics at the start of ibrutinib treatment are presented in Supplementary Table 1. The patients received a median of one other treatment (range, 1–5) before ibrutinib administration. The ORR was 69% (N=29), with 21 CR (50%) and eight PR (19%). Patients (N=29, 69.0%), who had previously received one line of treatment, were administered a median of 12 cycles of ibrutinib (range, 1–55), and they achieved an ORR of 72.4% (N=21), with 16 CR (55.1%) and five PR (17.2%). Furthermore, patients (N=13, 31.0%), previously treated with more than two lines of treatment, received a median of 14 cycles of ibrutinib (range, 3–23), and they achieved an ORR of 61.5% (N=8), with five CR (38.4%) and three PR (23%). The most common reasons for treatment discontinuation were disease progression (N=23, 54.8%) and severe AEs (N=5, 11.9%).

During a median follow-up of 46.0 months (95% CI, 1.9–80.5), the median PFS was 16.4 months (95% CI, 0.0–41.5), and the median OS was 50.1 months (95% CI, 9.7–90.5; Fig. 1A, B). The responders showed superior PFS (46.0 mo; 95% CI, 26.2–65.7 vs. 7.9 mo; 95% CI, 2.8–12.9; P=0.001) and OS (63.5 mo; 95% CI, 42.9–67.3 vs. 13.6 mo; 95% CI, 7.3–19.9; P=0.002), compared to poorly responding patients (Fig. 1C, D). There was no difference in PFS (P=0.730) or OS (P=0.544) according to the line of treatment (Fig. 1E, F). However, patients, who experienced disease progression within 24 months of front-line treatment (POD 24), exhibited inferior PFS (10.5 mo; 95% CI, 4.4–16.6 vs. 53.8 mo; 95% CI, 27.9–79.6; P=0.005) and OS (12.6 mo; 95% CI, 4.5–20.7 vs. NR; P=0.015) upon receiving salvage ibrutinib treatment (Fig. 1G, H).

Figure 1. Kaplan-Meier curve of progression-free survival (PFS) (A) and overall survival (OS) for salvage ibrutinib therapy (B). Comparison of PFS and OS of ibrutinib treatment according to response (C, D), line of treatment (E, F), and progression of disease within 24 months of front-line treatment (G, H). Assessment of PFS according to age (I), presence of splenomegaly (J), and simplified mantle cell lymphoma international prognostic index (sMIPI) (K) and OS according to age (L), presence of splenomegaly (M), and sMIPI (N).

Patients, who were older than 70 years at the initiation of ibrutinib treatment (7.9 mo; 95% CI, 1.3–14.4 vs. 36.6 mo; 95% CI, 9.8–63.5; P=0.004), or those with splenomegaly (9.9 mo; 95% CI, 0.4–19.4 vs. 46.0 mo; 95% CI, 10.3–81.6; P=0.022) or a high-risk sMIPI (5.0 mo; 95% CI, 2.3–7.7 vs. 37.4 mo; 95% CI, 29.4–45.4 vs. 63.5 mo; 95% CI, 0.0–152.8; P<0.001) exhibited inferior PFS with ibrutinib treatment (Fig. 1I–K). Moreover, patients older than 70 years (13.6 mo; 95% CI, 6.7–20.5 vs. 63.5 mo; 95% CI, 9.8–63.5; P=0.006) or those with splenomegaly (12.6 mo; 95% CI, 0.0–34.0 vs. not reached [NR]; P=0.006) or a high-risk sMIPI (9.9 mo; 95% CI, 3.4–16.4 vs. NR vs. NR; P<0.001) displayed poorer OS (Fig. 1L–N). In the univariate and multivariate analyses, the duration of ibrutinib treatment and sMIPI were significant factors, influencing both OS and PFS (Table 2).

Table 2 . Univariate and multivariate analyses for progressiong-free survival and overall survival with salvage ibrutinib treatment..

VariablesOverall survivalProgression-free survival
UnivariateMultivariateUnivariateMultivariate
HR (95% CI)PHR (95% CI)PHR (95% CI)PHR (95% CI)P
Age (≥70 yr)3.419 (1.340–8.726)0.0100.380 (0.106–1.358)0.1373.158 (1.395–7.146)0.0060.578 (0.201–1.661)0.308
sMIPI score (high score)5.325 (2.111–13.428)<0.0016.249 (2.067–18.892)0.0014.681 (2.086–10.501)<0.0012.874 (1.171–7.055)0.021
Splenomegaly3.303 (1.345–8.111)0.0091.983 (0.439–8.961)0.3742.338 (1.107–4.936)0.0261.373 (0.506–3.720)0.534
Anthracycline-contai- ning front-line treatment0.197 (0.075–0.516)0.0011.282 (0.354–4.647)0.7050.159 (0.063–0.402)<0.0010.273 (0.081–0.921)0.036
POD 24 of front-line treatment3.013 (1.190–7.625)0.0200.991 (0.242–4.070)0.9912.858 (1.336–6.117)0.0071.895 (0.737–4.876)0.185
Previous number of treatments (≥2 lines )0.730 (0.263–2.024)0.5461.230 (0.576–2.626)0.593
Underwent ASCT before ibrutinib treatment0.692 (0.230–2.086)0.5131.068 (0.468–2.439)0.876
Duration of ibrutinib >12 months0.023 (0.003–0.179)<0.0010.011 (0.001–0.110)<0.0010.041 (0.011–0.154)<0.0010.042 (0.010–0.180)<0.001
Grade 3/4 adverse events3.185 (1.203–8.433)0.0201.884 (0.612–5.798)0.2691.977 (0.789–4.953)0.146

Abbreviations: ASCT, autologous stem cell transplantation; POD 24, disease progression within 24 months; sMIPI, simplified mantle cell lymphoma international prognostic index..



Characteristics of the patients who experienced early relapse

An analysis was conducted to confirm the clinical markers, affecting early relapse during ibrutinib treatment (N=13, 30.9%). According to the independent samples t-test, only a high-risk sMIPI was correlated with early relapse (P=0.033, Table 3). Patients with early relapse had significantly poorer survival outcomes than those, who did not experience early relapse (12.6 mo; 95% CI, 7.9–17.3 vs. NR; P<0.001, Fig. 2A). The primary non-responders had exceptionally poor survival compared to secondary non-responders and consistent responders (5.9 mo; 95% CI, 0.7–11.1 vs. 30.7 mo; 95% CI, 1.2–60.2 vs. NR; P=0.002, Fig. 2B). In terms of cumulative incidence, patients who received BR as front-line treatment (P=0.004), those with high-risk sMIPI (P=0.010), and those older than 70 years (P=0.011) were strongly correlated with an increased cumulative incidence of early relapse (Fig. 2C–E). However, there was a weak correlation between early relapse, splenomegaly (P=0.084), and prior lines of treatment (P=0.720) (Fig. 2F, G).

Figure 2. Kaplan-Meier curves of overall survival according to early relapse (A) and type of response to salvage ibrutinib (B). Comparison of cumulative incidence of early relapse according to front-line treatment (C), simplified mantle cell lymphoma international prognostic index (D), age (E), presence of splenomegaly (F), and line of treat-ment of ibrutinib (G).

Table 3 . Comparison of patients with and without early disease relapse..

CharacteristicsEarly relapse (N=13)Late relapse or no relapse (N=29)P
Age (yr)Median (yr), range67 (51–77)67 (45–81)
<706 (46.2%)21 (72.4%)0.101
≥707 (53.8%)8 (27.6%)
GenderMale10 (76.9%)20 (69.0%)0.598
Female3 (23.1%)9 (31.0%)
ECOG-PS0–112 (92.3%)29 (100.0%)0.131
2–41 (7.7%)0 (0.0%)
sMIPI scoreLow (0–3)0 (0.0%)7 (24.1%)0.033
Intermediate (4–5)6 (46.2%)14 (48.3%)
High (6–11)7 (53.8%)8 (27.6%)
Splenomegaly (≥11 cm)Absence7 (53.8%)11 (37.9%)0.335
Present6 (46.2%)18 (62.1%)
Prior number of treatmentPrior 1 line9 (69.2%)20 (69.0%)0.986
Prior ≥2 lines4 (30.8%)9 (31.0%)
Front-line treatmentAnthracycline-based6 (46.2%)20 (69.0%)0.159
Bendamustine-based7 (53.8%)9 (31.0%)
ASCTReceived3 (23.1%)7 (24.1%)0.941
Not received10 (76.9%)22 (75.9%)
Ibrutinib dose adjustmentDose reduction1 (7.7%)5 (17.2%)0.414
No dose reduction12 (92.3%)24 (82.8%)
Ibrutinib discontinuationDiscontinuation13 (100.0%)24 (82.8%)0.111
No discontinuation0 (0.0%)5 (17.2%)

Abbreviations: ASCT, autologous stem cell transplantation; ECOG-PS, Eastern Cooperative Oncology Group Performance Status; sMIPI, simplified mantle cell lymphoma international prognostic index..



Post-ibrutinib survival outcomes

The median OS for patients, who discontinued ibrutinib treatment (N=37, 88.1%), was 9.8 months (95% CI, 0.6–19.0) (Fig. 3A). The ibrutinib responders had a better post-ibrutinib OS than the non-responders, but the difference was not statistically significant (20.0 mo; 95% CI, 0.0–43.0 vs. 5.7 mo; 95% CI, 0.0–13.2; P=0.101). The primary non-responders, who did not achieve disease control, had a median OS of only 2.6 months (95% CI, 0.0–10.4) after ibrutinib treatment.

Figure 3. Kaplan-Meier curves of overall survival for post-ibrutinib treatment (A). Comparison of overall survival according to participation in clinical trials (B). Swimmer plot of patients who received post-ibrutinib treatment (C).

Twenty-three patients (62.1%) received additional anti-cancer therapies after ibrutinib discontinuation, and the estimated median OS after ibrutinib was 12.2 months (95% CI, 2.1–22.2). Sixty-five percent of patients (N=15) received bortezomib or novel agents, such as lenalidomide and venetoclax, and 21.7% (N=5) participated in clinical trials. Although statistical significance was not achieved, the patients assigned to clinical trials had improved survival outcomes, compared to those, who received conventional chemotherapy (P=0.458, Fig. 3B). In terms of post-ibrutinib survival, the patients, who experienced early relapse, generally had an inferior OS (5.4 mo; 95% CI, 0.0–12.7 vs. 24.8 mo; 95% CI, 15.7–37.2; P=0.012) (Fig. 3C).

Adverse events with ibrutinib treatment

During ibrutinib treatment, various hematologic and non-hematologic AEs were observed, but most were reported as grade 1 or 2 and were manageable. Six patients (14.3%) received a reduced ibrutinib dose, and five patients (11.9%) discontinued ibrutinib treatment due to toxicities (Supplementary Table 2). All grades of thrombocytopenia accounted for 11.9% of the hematologic AEs (N=5). In particular, grade ≥3 thrombocytopenia accounted for 7.1% (N=3). The incidence of all grades of anemia and neutropenia was 7.1%, and less than 5% had a grade ≥3. The most common non-hematologic AE was skin rash (N=20, 47.6%), and only four patients (9.5%) presented with a grade ≥3. Grade ≥3 fatigue and diarrhea were observed in approximately 9.5% (N=4) and 7.1% (N=3), respectively. Three patients (7.1%) had grade 1 or 2 atrial fibrillation, and all were adequately managed through medical intervention. Of the five patients who discontinued ibrutinib, one ceased treatment due to a hematologic AE (grade 3 thrombocytopenia), while the remaining four stopped due to non-hematologic AEs (two patients with grade 3 diarrhea, one with grade 3 rash, and one with grade 3 fatigue). Among the consistent responders older than 70 years, those who were maintained on ibrutinib treatment with well-managed severe AEs had a more favorable PFS (36.6 mo; 95% CI, 12.3–60.9 vs. 11.4 mo; 95% CI, 0.0–23.1; P=0.234) and OS (NR vs. 11.4 mo; 95% CI, 0.0–23.1; P=0.062) (Supplementary Fig. 1A).

DISCUSSION

This study evaluated the efficacy and survival outcomes of ibrutinib treatment, based on real-world clinical experience. According to previous studies (Table 4) [5-14],

Table 4 . Summary of previous studies on ibrutinib therapy in relapsed or refractory mantle cell lymphoma..

StudyPt. numberFront-line Tx.Refractory to front-line Tx.Upfront ASCTPrior Tx.ORRPFS (median)OS (median)Ref
Phase 2111R-containing therapy (N=99, 89%)50 (45%)8 (13%)3 (1–5)67% (CR, 23%)13 mo22.5 mo[5, 6]
Phase 3280R-containing therapy (N=280, 100%)36 (26%)N/A2 (1–9)72% (CR, 19%)14.6 moNot reached[7]
Retrospective97Intensive induction therapy (N=52, 54%) (R±hyper-CVAD, R-maxi CHOP)7 (7%)38 (39%)2 (1–8)65% (CR, 33%)15 mo22 mo[8]
Phase 216R-containing therapy (N=16, 100%)1 (6.2%)N/A2.5 (1–4)87.5% (CR, 12.5%)N/AN/A[9]
Retrospective33R-CHOP (N=33)N/A6 (18.1%)

1 (33.3%).

2 (36.4%).

≥3 (30.3%).

64% (CR, 15%)27.4 mo35.1 mo[10]
Retrospective65R-CHOP, R-bendamustine and R-hyper-CVAD (87%)N/AN/A2 (1–6)N/A12.0 mo18.5 mo[11]
Retrospective88

R±CHOP (N=63).

R±hyper-CVAD (N=13).

BR (N=6).

VR-CAP (N=3).

CVP/bendamustine (N=3).

N/A5 (5.6%)1 (1–6)64.8% (CR, N/A)20.8 mo2-yr OS rate 79.5%[12]
Retrospective211

R-CHOP/mini-CHOP (N=70, 33%).

Cytarabine-based regimen (N=60, 28%).

R-bendamustine (N=45, 21%).

VR-CAP (N=5, 2%).

Others (N=31, 15%).

38 (18%)53 (25%)169% (CR, 27%)17.8 mo23.9 mo[13]
Retrospective77

CHOP/CHOP-like (N=42, 54%).

Cytarabine-based regimen (N=25, 33%).

Non-anthracycline regimen (N=10, 13%).

15 (20%)19 (25%)

1 (29%).

2 (30%).

≥3 (41%).

66%.

(CR, 31%).

10.3 mo23.1 mo[14]
Retrospective42

R-CHOP (N=18, 43%).

R-hyper-CVAD (N=8, 19%).

R-bendamustine (N=16, 38%).

20 (47.6%)6 (14.3%)1 (1–5)69% (CR, 50%)19.2 mo50.1 moCurrent study

Abbreviations: ASCT, autologous stem cell transplantation; BR, rituximab, bendamustine; CHOP, cyclophosphamide, doxorubicin, vincristine, prednisolone; CVP, cyclophosphamide, vincristine, prednisone; Hyper-CVAD, hyperfractionated cyclophosphamide, vincristine, doxorubicin dexamethasone; LOT, line of therapy; N/A, non-available; ORR, objective response rate; OS, overall survival; PFS, progression-free survival; Pt., patient; R, rituximab; Tx., treatment; VR-CAP, bortezomib, rituximab, cyclophosphamide, doxorubicin, prednisolone..



Based on the CAN3001 study, which reported the long-term outcomes of salvage ibrutinib treatment with 10 years of follow-up [20], severe treatment-related AEs (TRAEs) were generally observed within one year of ibrutinib initiation, but it decreased annually thereafter. The AEs reported in this study were comparable to those, documented in previous studies, in terms of the type, timing of occurrence, and severity. This underscores that the prompt and proactive management of AEs during ibrutinib treatment alleviates the patient’s symptoms and enhances survival outcomes. The patients, who received active toxicity management along with ibrutinib, had a stable treatment course and improved clinical outcomes despite AEs. Thus, to prevent treatment interruption and achieve favorable outcomes, acute and chronic AEs, associated with ibrutinib therapy, should be recognized, monitored, and managed.

In this study, ibrutinib monotherapy resulted in an ORR of approximately 70%, which was similar to or higher than the response rates, reported in other studies (Table 4). Ibrutinib was more efficacious in patients, older than 70 years, who received anthracycline-containing front-line treatment, than in those, who received bendamustin-based front-line treatment (Supplementary Fig. 1B). Patients, treated with a bendamustine-containing regimen, presented with unfavorable baseline characteristics, including age >70 years, elevated WBC count, BM involvement, and high-risk sMIPI at the time of diagnosis (Table 1). Based on this, less intensive chemotherapy is preferred as the front-line treatment for patients aged 70 or older as they have poor general health conditions or comorbidities. It was assumed that prior bendamustine exposure led to poor subsequent ibrutinib treatment outcomes, as observed in other studies [10]. In cases where POD 24 occurred, regardless of the front-line treatment, favorable survival outcomes were not guaranteed, even with ibrutinib treatment. In line with previous studies [20, 21], POD 24 persisted as an unfavorable prognostic indicator in the targeted agent era.

TP53 mutation is a well-known biomarker, that predicts poor outcomes with conventional treatments in both newly diagnosed and RR-MCL patients. According to several retrospective studies [15, 22], the incidence of TP53 mutations was approximately 14%, and patients with TP53 mutations had lower response rates (55% vs. 70%) as well as shorter PFS (4 mo vs. 12 mo) and OS (10 mo vs. 34 mo) than patients with wild-type TP53. Unfortunately, this study was unable to include data on TP53 mutation status due to limited tissue samples and the lack of patient consent. While the presence of TP53 mutations impacts the prognosis of MCL, performing NGS on all MCL patients was not cost-effective in a real-world setting since only 14% were found to have TP53 mutations. Instead, Eskelund et al. [23] suggested specific clinical factors, related to TP53 mutation. These include a blastoid morphology, high-risk MIPI, and Ki67 >30%. In cases, where determining the TP53 mutation status is challenging, establishing a treatment plan, based on the implied factors, and defining the clinical signs of TP53 mutations are essential.

In this study, early disease relapse was defined as disease relapse within 12 months of starting ibrutinib treatment since the reported median PFS of ibrutinib was 12.6 months (95% CI, 10.3–16.6). Patients with early relapse exhibited significantly shorter PFS than those, who did not experience early relapse (5.4 mo vs. 24.8 mo). High-risk sMIPI scores were correlated with early disease relapse. Unfortunately, these patients were unsuitable for any subsequent treatment due to rapid disease deterioration, and the post-ibrutinib survival outcomes were poor. Establishing optimal treatment strategies for patients, who were refractory to ibrutinib, remains challenging. Allogeneic SCT has demonstrated long-term disease control through the graft-to-host effect, but it is limited to patients with favorable general conditions for withstanding intensive induction therapy and life-threatening complications. Pirtobrutinib, a novel non-covalent BKT inhibitor, achieved an ORR of 58%. Patients with RR-MCL, who received a median of three prior treatment modalities, including ibrutinib, responded for 21.6 months [24]. Moreover, brexucabtagene autoleucel (brex-cel), an anti-CD19 CAR-T cell therapy, achieved an ORR of 93% and CR rate of 67% [16, 17]. After 12 months, 57% of patients remained in remission. The lisocaltagene maraleucel (liso-cel) was associated with a low incidence of grade ≥3 TRAEs, and it demonstrated promising clinical activity in a phase 1 study [25]. Thus, novel therapeutic approaches, including targeted agents and immunocellular therapies, should be utilized in the management of patients, experiencing early disease relapse. Additionally, the risk stratification system should be continuously validated and adjusted to improve treatment outcomes.

In conclusion, this study demonstrated the efficacy and safety of salvage ibrutinib in patients with RR-MCL, and the patients, who potentially benefit from salvage ibrutinib therapy, were identified by verifying prognostic factors. Favorable survival outcomes were more frequent among patients with an objective response or those, who remained in disease remission with continuous ibrutinib treatment. To maximize the benefits of ibrutinib treatment, the capacity of the patient to endure TRAEs and the specific characteristics of their disease should be considered. Since patients, who experience early disease relapse within 12 months, have poorer survival outcomes, groundbreaking treatment strategies should be identified.

Authors’ Disclosures of Potential Conflicts of Interest

No potential conflicts of interest relevant to this article were reported.

Fig 1.

Figure 1.Kaplan-Meier curve of progression-free survival (PFS) (A) and overall survival (OS) for salvage ibrutinib therapy (B). Comparison of PFS and OS of ibrutinib treatment according to response (C, D), line of treatment (E, F), and progression of disease within 24 months of front-line treatment (G, H). Assessment of PFS according to age (I), presence of splenomegaly (J), and simplified mantle cell lymphoma international prognostic index (sMIPI) (K) and OS according to age (L), presence of splenomegaly (M), and sMIPI (N).
Blood Research 2023; 58: 208-220https://doi.org/10.5045/br.2023.2023208

Fig 2.

Figure 2.Kaplan-Meier curves of overall survival according to early relapse (A) and type of response to salvage ibrutinib (B). Comparison of cumulative incidence of early relapse according to front-line treatment (C), simplified mantle cell lymphoma international prognostic index (D), age (E), presence of splenomegaly (F), and line of treat-ment of ibrutinib (G).
Blood Research 2023; 58: 208-220https://doi.org/10.5045/br.2023.2023208

Fig 3.

Figure 3.Kaplan-Meier curves of overall survival for post-ibrutinib treatment (A). Comparison of overall survival according to participation in clinical trials (B). Swimmer plot of patients who received post-ibrutinib treatment (C).
Blood Research 2023; 58: 208-220https://doi.org/10.5045/br.2023.2023208

Table 1 . Baseline demographics and disease characteristics at diagnosis..

CharacteristicsTotal
(N=42)
BR
(N=16)
Anthracycline-based regimen (N=26)P
Age (yr)Median (range)62 (38–78)63 (50–78)61 (39–70)
<6017 (40.5%)2 (12.5%)15 (57.7%)0.004
≥6025 (59.5%)14 (87.5%)11 (42.3%)
GenderMale30 (71.4%)11 (68.7%)19 (73.0%)0.763
Female12 (28.6%)5 (31.2%)7 (27.0%)
ECOG-PS0–139 (92.9%)14 (87.5%)25 (96.2%)0.290
2–43 (7.1%)2 (12.5%)1 (3.8%)
B symptomAbsence37 (88.1%)13 (81.2%)24 (92.3%)0.283
Present5 (11.9%)3 (18.7%)2 (7.7%)
LeukocytosisAbsence31 (73.8%)8 (50.0%)23 (88.5%)0.006
Present11 (26.2%)8 (50.0%)3 (11.5%)
AnemiaAbsence33 (78.6%)9 (56.2%)24 (92.3%)0.006
Present9 (21.4%)7 (43.7%)2 (7.7%)
ThrombocytopeniaAbsence36 (85.7%)11 (68.7%)25 (96.2%)0.014
Present6 (14.3%)5 (31.2%)1 (3.8%)
Serum LDHAbsence23 (54.8%)7 (43.7%)16 (61.5%)0.261
Elevated19 (45.2%)9 (56.2%)10 (38.5%)
Serum β-2 microglobulinAbsence23 (54.8%)5 (31.2%)18 (69.2%)0.016
Elevated19 (45.2%)11 (68.7%)8 (30.8%)
Bone marrow involvementAbsence13 (31.0%)2 (12.5%)11 (42.3%)0.042
Present29 (69.0%)14 (87.5%)15 (57.7%)
Number of extra-nodal involvementLess than one11 (26.2%)2 (12.5%)9 (34.6%)0.113
More than two31 (73.8%)14 (87.5%)17 (65.4%)
Bulky (≥7 cm) massAbsence33 (78.6%)10 (62.5%)23 (88.5%)0.046
Present9 (21.4%)6 (37.5%)3 (11.5%)
PathologyClassic type41 (97.6%)16 (100.0%)25 (96.2%)0.427
Pleomorphic/blastoid type1 (2.4%)0 (0.0%)1 (3.8%)
Ki-67<30%33 (78.6%)12 (75.0%)21 (80.8%)0.658
≥30%9 (21.4%)4 (25.0%)5 (19.2%)
Ann Arbor stageI/II5 (11.9%)1 (6.3%)4 (15.4%)0.375
III/IV37 (88.1%)15 (93.7%)22 (84.6%)
sMIPI scoreLow (0–3)19 (45.2%)3 (18.7%)16 (61.5%)0.002
Intermediate (4–5)18 (42.9%)8 (50.0%)10 (38.5%)
High (6–11)5 (11.9%)5 (31.3%)0 (0.0%)

Abbreviations: ASCT, autologous stem cell transplantation; BR, bendamustine plus rituximab; ECOG-PS, Eastern Cooperative Oncology Group Performance Status; LDH, lactate dehydrogenase; sMIPI, simplified mantle cell lymphoma international prognostic index..


Table 2 . Univariate and multivariate analyses for progressiong-free survival and overall survival with salvage ibrutinib treatment..

VariablesOverall survivalProgression-free survival
UnivariateMultivariateUnivariateMultivariate
HR (95% CI)PHR (95% CI)PHR (95% CI)PHR (95% CI)P
Age (≥70 yr)3.419 (1.340–8.726)0.0100.380 (0.106–1.358)0.1373.158 (1.395–7.146)0.0060.578 (0.201–1.661)0.308
sMIPI score (high score)5.325 (2.111–13.428)<0.0016.249 (2.067–18.892)0.0014.681 (2.086–10.501)<0.0012.874 (1.171–7.055)0.021
Splenomegaly3.303 (1.345–8.111)0.0091.983 (0.439–8.961)0.3742.338 (1.107–4.936)0.0261.373 (0.506–3.720)0.534
Anthracycline-contai- ning front-line treatment0.197 (0.075–0.516)0.0011.282 (0.354–4.647)0.7050.159 (0.063–0.402)<0.0010.273 (0.081–0.921)0.036
POD 24 of front-line treatment3.013 (1.190–7.625)0.0200.991 (0.242–4.070)0.9912.858 (1.336–6.117)0.0071.895 (0.737–4.876)0.185
Previous number of treatments (≥2 lines )0.730 (0.263–2.024)0.5461.230 (0.576–2.626)0.593
Underwent ASCT before ibrutinib treatment0.692 (0.230–2.086)0.5131.068 (0.468–2.439)0.876
Duration of ibrutinib >12 months0.023 (0.003–0.179)<0.0010.011 (0.001–0.110)<0.0010.041 (0.011–0.154)<0.0010.042 (0.010–0.180)<0.001
Grade 3/4 adverse events3.185 (1.203–8.433)0.0201.884 (0.612–5.798)0.2691.977 (0.789–4.953)0.146

Abbreviations: ASCT, autologous stem cell transplantation; POD 24, disease progression within 24 months; sMIPI, simplified mantle cell lymphoma international prognostic index..


Table 3 . Comparison of patients with and without early disease relapse..

CharacteristicsEarly relapse (N=13)Late relapse or no relapse (N=29)P
Age (yr)Median (yr), range67 (51–77)67 (45–81)
<706 (46.2%)21 (72.4%)0.101
≥707 (53.8%)8 (27.6%)
GenderMale10 (76.9%)20 (69.0%)0.598
Female3 (23.1%)9 (31.0%)
ECOG-PS0–112 (92.3%)29 (100.0%)0.131
2–41 (7.7%)0 (0.0%)
sMIPI scoreLow (0–3)0 (0.0%)7 (24.1%)0.033
Intermediate (4–5)6 (46.2%)14 (48.3%)
High (6–11)7 (53.8%)8 (27.6%)
Splenomegaly (≥11 cm)Absence7 (53.8%)11 (37.9%)0.335
Present6 (46.2%)18 (62.1%)
Prior number of treatmentPrior 1 line9 (69.2%)20 (69.0%)0.986
Prior ≥2 lines4 (30.8%)9 (31.0%)
Front-line treatmentAnthracycline-based6 (46.2%)20 (69.0%)0.159
Bendamustine-based7 (53.8%)9 (31.0%)
ASCTReceived3 (23.1%)7 (24.1%)0.941
Not received10 (76.9%)22 (75.9%)
Ibrutinib dose adjustmentDose reduction1 (7.7%)5 (17.2%)0.414
No dose reduction12 (92.3%)24 (82.8%)
Ibrutinib discontinuationDiscontinuation13 (100.0%)24 (82.8%)0.111
No discontinuation0 (0.0%)5 (17.2%)

Abbreviations: ASCT, autologous stem cell transplantation; ECOG-PS, Eastern Cooperative Oncology Group Performance Status; sMIPI, simplified mantle cell lymphoma international prognostic index..


Table 4 . Summary of previous studies on ibrutinib therapy in relapsed or refractory mantle cell lymphoma..

StudyPt. numberFront-line Tx.Refractory to front-line Tx.Upfront ASCTPrior Tx.ORRPFS (median)OS (median)Ref
Phase 2111R-containing therapy (N=99, 89%)50 (45%)8 (13%)3 (1–5)67% (CR, 23%)13 mo22.5 mo[5, 6]
Phase 3280R-containing therapy (N=280, 100%)36 (26%)N/A2 (1–9)72% (CR, 19%)14.6 moNot reached[7]
Retrospective97Intensive induction therapy (N=52, 54%) (R±hyper-CVAD, R-maxi CHOP)7 (7%)38 (39%)2 (1–8)65% (CR, 33%)15 mo22 mo[8]
Phase 216R-containing therapy (N=16, 100%)1 (6.2%)N/A2.5 (1–4)87.5% (CR, 12.5%)N/AN/A[9]
Retrospective33R-CHOP (N=33)N/A6 (18.1%)

1 (33.3%).

2 (36.4%).

≥3 (30.3%).

64% (CR, 15%)27.4 mo35.1 mo[10]
Retrospective65R-CHOP, R-bendamustine and R-hyper-CVAD (87%)N/AN/A2 (1–6)N/A12.0 mo18.5 mo[11]
Retrospective88

R±CHOP (N=63).

R±hyper-CVAD (N=13).

BR (N=6).

VR-CAP (N=3).

CVP/bendamustine (N=3).

N/A5 (5.6%)1 (1–6)64.8% (CR, N/A)20.8 mo2-yr OS rate 79.5%[12]
Retrospective211

R-CHOP/mini-CHOP (N=70, 33%).

Cytarabine-based regimen (N=60, 28%).

R-bendamustine (N=45, 21%).

VR-CAP (N=5, 2%).

Others (N=31, 15%).

38 (18%)53 (25%)169% (CR, 27%)17.8 mo23.9 mo[13]
Retrospective77

CHOP/CHOP-like (N=42, 54%).

Cytarabine-based regimen (N=25, 33%).

Non-anthracycline regimen (N=10, 13%).

15 (20%)19 (25%)

1 (29%).

2 (30%).

≥3 (41%).

66%.

(CR, 31%).

10.3 mo23.1 mo[14]
Retrospective42

R-CHOP (N=18, 43%).

R-hyper-CVAD (N=8, 19%).

R-bendamustine (N=16, 38%).

20 (47.6%)6 (14.3%)1 (1–5)69% (CR, 50%)19.2 mo50.1 moCurrent study

Abbreviations: ASCT, autologous stem cell transplantation; BR, rituximab, bendamustine; CHOP, cyclophosphamide, doxorubicin, vincristine, prednisolone; CVP, cyclophosphamide, vincristine, prednisone; Hyper-CVAD, hyperfractionated cyclophosphamide, vincristine, doxorubicin dexamethasone; LOT, line of therapy; N/A, non-available; ORR, objective response rate; OS, overall survival; PFS, progression-free survival; Pt., patient; R, rituximab; Tx., treatment; VR-CAP, bortezomib, rituximab, cyclophosphamide, doxorubicin, prednisolone..


References

  1. Armitage JO, Longo DL. Mantle-cell lymphoma. N Engl J Med 2022;386:2495-506.
    Pubmed CrossRef
  2. Lee H, Park HJ, Park EH, et al. Nationwide statistical analysis of lymphoid malignancies in Korea. Cancer Res Treat 2018;50:222-38.
    Pubmed KoreaMed CrossRef
  3. Merolle MI, Ahmed M, Nomie K, Wang ML. The B cell receptor signaling pathway in mantle cell lymphoma. Oncotarget 2018;9:25332-41.
    Pubmed KoreaMed CrossRef
  4. Gu D, Tang H, Wu J, Li J, Miao Y. Targeting Bruton tyrosine kinase using noncovalent inhibitors in B cell malignancies. J Hematol Oncol 2021;14:40.
    Pubmed KoreaMed CrossRef
  5. Wang ML, Rule S, Martin P, et al. Targeting BTK with ibrutinib in relapsed or refractory mantle-cell lymphoma. N Engl J Med 2013;369:507-16.
    Pubmed KoreaMed CrossRef
  6. Wang ML, Blum KA, Martin P, et al. Long-term follow-up of MCL patients treated with single-agent ibrutinib: updated safety and efficacy results. Blood 2015;126:739-45.
    Pubmed KoreaMed CrossRef
  7. Dreyling M, Jurczak W, Jerkeman M, et al. Ibrutinib versus temsirolimus in patients with relapsed or refractory mantle-cell lymphoma: an international, randomised, open-label, phase 3 study. Lancet 2016;387:770-8.
    Pubmed CrossRef
  8. Epperla N, Hamadani M, Cashen AF, et al. Predictive factors and outcomes for ibrutinib therapy in relapsed/refractory mantle cell lymphoma-a "real world" study. Hematol Oncol 2017;35:528-35.
    Pubmed CrossRef
  9. Maruyama D, Nagai H, Fukuhara N, et al. Efficacy and safety of ibrutinib in Japanese patients with relapsed or refractory mantle cell lymphoma. Cancer Sci 2016;107:1785-90.
    Pubmed KoreaMed CrossRef
  10. Jeon YW, Yoon S, Min GJ, et al. Clinical outcomes for ibrutinib in relapsed or refractory mantle cell lymphoma in real-world experience. Cancer Med 2019;8:6860-70.
    Pubmed KoreaMed CrossRef
  11. Tucker D, Morley N, MacLean P, et al. The 5-year follow-up of a real-world observational study of patients in the United Kingdom and Ireland receiving ibrutinib for relapsed/refractory mantle cell lymphoma. Br J Haematol 2021;192:1035-8.
    Pubmed CrossRef
  12. Yi JH, Kim SJ, Yoon DH, et al. Real-world outcomes of ibrutinib therapy in Korean patients with relapsed or refractory mantle cell lymphoma: a multicenter, retrospective analysis. Cancer Commun (Lond) 2021;41:275-8.
    Pubmed KoreaMed CrossRef
  13. McCulloch R, Lewis D, Crosbie N, et al. Ibrutinib for mantle cell lymphoma at first relapse: a United Kingdom real-world analysis of outcomes in 211 patients. Br J Haematol 2021;193:290-8.
    Pubmed CrossRef
  14. Obr A, Benesova K, Janikova A, et al. Ibrutinib in mantle cell lymphoma: a real-world retrospective multi-center analysis of 77 patients treated in the Czech Republic. Ann Hematol 2023;102:107-15.
    Pubmed KoreaMed CrossRef
  15. Rule S, Dreyling M, Goy A, et al. Ibrutinib for the treatment of relapsed/refractory mantle cell lymphoma: extended 3.5-year follow up from a pooled analysis. Haematologica 2019;104:e211-4.
    Pubmed KoreaMed CrossRef
  16. Wang M, Munoz J, Goy A, et al. KTE-X19 CAR T-cell therapy in relapsed or refractory mantle-cell lymphoma. N Engl J Med 2020;382:1331-42.
    Pubmed KoreaMed CrossRef
  17. Wang M, Munoz J, Goy A, et al. Three-year follow-up of KTE-X19 in patients with relapsed/refractory mantle cell lymphoma, including high-risk subgroups, in the ZUMA-2 study. J Clin Oncol 2023;41:555-67.
    Pubmed KoreaMed CrossRef
  18. Swerdlow SH, Campo E, Pileri SA, et al. The 2016 revision of the World Health Organization classification of lymphoid neoplasms. Blood 2016;127:2375-90.
    Pubmed KoreaMed CrossRef
  19. Cheson BD, Fisher RI, Barrington SF, et al. Recommendations for initial evaluation, staging, and response assessment of Hodgkin and non-Hodgkin lymphoma: the Lugano classification. J Clin Oncol 2014;32:3059-68.
    Pubmed KoreaMed CrossRef
  20. Dreyling M, Goy A, Hess G, et al. Long-term outcomes with ibrutinib treatment for patients with relapsed/refractory mantle cell lymphoma: a pooled analysis of 3 clinical trials with nearly 10 years of follow-up. Hemasphere 2022;6:e712.
    Pubmed KoreaMed CrossRef
  21. Bond DA, Switchenko JM, Maddocks KJ, et al. Outcomes following early relapse in patients with mantle cell lymphoma. Blood (ASH Annual Meeting Abstracts) 2019;134(Suppl 1):753.
    CrossRef
  22. Lew TE, Minson A, Dickinson M, et al. Treatment approaches for patients with TP53-mutated mantle cell lymphoma. Lancet Haematol 2023;10:e142-54.
    Pubmed CrossRef
  23. Eskelund CW, Dahl C, Hansen JW, et al. TP53 mutations identify younger mantle cell lymphoma patients who do not benefit from intensive chemoimmunotherapy. Blood 2017;130:1903-10.
    Pubmed CrossRef
  24. Wang ML, Jurczak W, Zinzani PL, et al. Pirtobrutinib in covalent bruton tyrosine kinase inhibitor pretreated mantle-cell lymphoma. J Clin Oncol 2023;41:3988-97.
    Pubmed KoreaMed CrossRef
  25. Palomba ML, Gordon LI, Siddiqi T, et al. Safety and preliminary efficacy in patients with relapsed/refractory mantle cell lymphoma receiving lisocabtagene maraleucel in transcend NHL 001. Blood (ASH Annual Meeting Abstracts) 2020;136(Suppl 1):10-1.
    CrossRef
Blood Res
Volume 59 2024

Supplementary File

Stats or Metrics

Share this article on

  • line

Related articles in BR

Blood Research

pISSN 2287-979X
eISSN 2288-0011
qr-code Download