Next Article in Journal
Solid Pseudopapillary Neoplasm of the Pancreas: A Comprehensive Review Focusing on the Role of Endoscopic Ultrasound-Guided Radiofrequency Ablation as an Alternative Treatment
Previous Article in Journal
Role of Human Microbiome in Development and Management of Head and Neck Squamous Cell Carcinoma
Previous Article in Special Issue
Single-Agent and Associated Therapies with Monoclonal Antibodies: What About Follicular Lymphoma?
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Next-Generation Therapies in Mantle Cell Lymphoma (MCL): The Evolving Landscape in Treatment of Relapse/Refractory After CAR-T Cells

1
Division of Hematology, Santa Croce e Carle Hospital, via Michele Coppino 26, 12100 Cuneo, Italy
2
Transfusion Medicine and Blood Establishment, Santa Croce e Carle Hospital, via Michele Coppino 26, 12100 Cuneo, Italy
3
Division of Hematology, Santi Antonio e Biagio e Cesare Arrigo Hospital, Spalto Marengo 43, 15121 Alessandria, Italy
4
Department of Translational Medicine, University of Eastern Piedmont, via Paolo Solaroli 17, 28100 Novara, Italy
5
Laboratory of Blood Tumor Immunology, Molecular Biotechnology Center “Guido Tarone”, University of Torino, via Nizza 52, 10126 Torino, Italy
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Cancers 2025, 17(13), 2239; https://doi.org/10.3390/cancers17132239
Submission received: 3 May 2025 / Revised: 6 June 2025 / Accepted: 27 June 2025 / Published: 3 July 2025
(This article belongs to the Special Issue Monoclonal Antibodies in Lymphoma)

Simple Summary

Mantle cell lymphoma (MCL) patients who were Relapsed/Refractory (R/R) after chimeric antigen receptor (CAR)-T cells or ineligible for that treatment had an unfavorable prognosis. This review aims to make an overview on the novel treatment emerging in this tough setting, such as new non-covalent BTKi molecules and novel monoclonal antibodies (MABs), including Antibody drugs conjugate (ADC) and T-cell-engaging (TCE) products.

Abstract

Mantle cell lymphoma (MCL) is a biological and clinical heterogeneous disease, ranging from more indolent subtypes to highly aggressive ones with a dismal prognosis. Bruton’s tyrosine kinase inhibitors (BTKis), such as ibrutinib, ameliorated the outcome of relapse/refractory (R/R) patients, but responses are usually quite short, requiring further treatments. In this setting, immunotherapy was demonstrated to have a key role, first with chimeric antigen receptor (CAR)-T cells, which completely changed the prognosis of these patients, showing a high rate of and long-lasting responses. However, many cases do not yet respond or relapse after CAR-T cells, and a subset of patients are not eligible for CAR-T cells in the first place. In this particular setting of MCL patients, next-generation therapies are emerging, such as new non-covalent BTKi molecules and novel monoclonal antibodies (MABs), which represent a heterogeneous group of agents, including Antibody-drug conjugate (ADC) and T-cell-engaging (TCE) products. The impact of these agents is expected to be meaningful firstly on relapse/refractory disease and subsequently also in early treatment lines. The aim of this review is to explore the main therapeutic strategies emerging in the context of MCL patients who failed or are not eligible for CAR-T-cells treatment, focusing on the most relevant therapeutic drugs, especially on the different monoclonal antibodies.

1. Introduction

Mantle cell lymphoma (MCL) is a rare but heterogeneous subtype of non-Hodgkin lymphoma (NHL) with a prognosis ranging from very favorable in indolent cases to a poor prognosis for more aggressive variants [1]. Patients usually present with advanced-stage disease, with common extranodal involvement, including infiltration of bone marrow (53–82%), blood (50%), liver (25%), and the gastrointestinal tract (20–60%) [2,3].
Biologically, the pathognomonic signature is represented by cyclin D1 translocation and sometimes cyclin D2 or D3 translocations [4]. Clinical heterogeneity reflects partially a different cell of origin (COO): MCL can develop either from naïve-like cells and show a higher risk of progression or from memory-like cells of the adaptive immune system and present an indolent clinical course [5]. However, the most important regulator of proliferation in MCL is B-cell receptor (BCR) signaling [4,5]. According to different clinical behaviors and different COOs, the ICC/WHO 2022 update of lymphoid malignancies identified in MCL two distinct categories: nodal and non-nodal [4,6]. Nodal MCL (80–90% of cases) is characterized by unmutated immunoglobulin heavy-chain variable-region genes (IGHVs), Sex-Determining Region Y-Box 11 (SOX11) overexpression, COO by naïve-like cells, and generally more aggressive clinical behavior. Non-nodal leukemic MCL (10–20% of cases) typically displays mutated IGHV, SOX11 negativity, and COO from memory-like cells and presents with indolent biological behavior [4,6].
Histologically, three subtypes can be described: the “classical” MCL subtype and the pleomorphic and blastoid variants, with the variants displaying usually a more aggressive course and worse prognosis [3,4,6,7].
MCL has historically been considered an incurable disease, and the goal of management was often to prolong survival and reduce disease-related symptoms, in particular, in the relapsed/refractory (R/R) setting. In the last decades, the therapeutic landscape of MCL has been a topic of great interest, and molecular target validation has led to critical translational insights into novel therapies. Recently, results coming from large international trials have completely revolutionized both the frontline and R/R therapeutic approaches.

2. Prognostic Factors

Many clinical, serological, and biological factors are well known to be associated with worse prognosis in MCL. The MCL International Prognostic Index (MIPI) is based on four easy clinical factors—age, performance status, LDH, and leukocyte count—and demonstrated in many series high prognostic value [8]. In the MIPI score, each parameter has a different value, and it is able to stratify patients into low- (0–3 points, 44% of patients), intermediate- (4–5 points, 35% of cases), and high-risk (6–11 points, 21% of patients) groups, with statistically different median overall survivals: not reached vs. 51 months vs. 29 months, in the low- vs. intermediate- vs. high-risk group, respectively. There are other easy clinical features known to have prognostic value, such as advanced stage; splenomegaly; extranodal involvement; and the presence of B symptoms, anemia, or an elevated serum level of β2-microglobulin.
The MIPI score has been modified and implemented in different ways during the years. For example, the MIPI-c score has included the immunohistochemical determination of Ki-67 expression into the MIPI index, identifying in patients with a Ki-67 higher than 30% a worse prognostic group [9].
The proliferation rate, expressed as Ki-67 expression, and blastoid morphology have also proved reliable in the identification of a high-risk biology subgroup with significantly shorter survival and worse response to intensive chemotherapy regimens, with the particular relevance of the deletion of the short arm of chromosome 17 (del(17p)) or TP53 mutations, which have independent prognostic value in MCL patients [10]; this has been translated into the new lymphoma classification systems, whereby investigating the Ki67 proliferation index and TP53 status is mandatory at MCL diagnosis [6].
Furthermore, the minimal residual disease (MRD) status after treatment showed in many clinical trials a high prognostic value [11,12], but the application of MRD in clinical practice is still limited; also, the impact of MRD in the era of new targeted treatments, such as Bruton’s tyrosine kinase inhibitors (BTKis), remains a matter of open debate, particularly because BTKis are administered continuously until disease progression.
Recently, applying the whole-exome sequencing technique, four different biological clusters have been identified, with different outcomes.
Some additional data on new genomic analysis techniques, like optical genome mapping, are emerging, suggesting an impact of the burden of chromosomal aberration on patient prognosis, and NGS methodics are confirming the negative impact of TP53 mutations [13,14,15].
However, this stratification has not entered routine clinical practice yet.

3. New Milestones in MCL Treatment

Up to a few years ago, the standard of care in frontline treatment of MCL in young fit patients included an induction therapy with anthracycline and platinum-based chemotherapy regimens, followed by autologous stem cell transplantation (ASCT) consolidation for patients in complete remission (CR) [11,16]. In addition, a maintenance phase based on the anti-CD20 monoclonal antibody rituximab was demonstrated to improve survival rates if compared to observation alone [17]. As for older and/or unfit patients, backbone treatment with bendamustine-based chemoimmunotherapy followed by rituximab maintenance remains the standard of care [18,19]. An alternative chemoimmunotherapy regimen that could be considered in this setting is VR-CAP (Bortezomib, rituximab, cyclophosphamide, doxorubicin, and prednisone), which demonstrated high efficacy and a favorable safety profile in frontline MCL treatment [20] In relapsed MCL, monotherapies with BTKi, first of all Ibrutinib, have become the preferred salvage treatments in the second-line setting, based on their superior efficacy compared to conventional chemotherapy or other targeted agents [21,22,23]. Further than ibrutinib, other second-generation selective BTK inhibitors have been investigated alone and in combination in the setting of R/R MCL. First of all, Acalabrutinib is a second-generation, highly selective, and potent BTK inhibitor approved for the treatment of patients with R/R MCL who have received ≥1 prior therapies. It has been investigated as a single agent in R/R MCL in a large phase II trial (ACE-LY-004), demonstrating an ORR of 81.5%, with CR 47.6% [24,25]. Secondly, Zanubrutinib, which, like acalabrutinib, has been designed to overcome the limitation of the first-generation ibrutinib by delivering important and continuous inhibition of the BTK protein, thus optimizing bioavailability, half-life, and selectivity [26,27,28,29]. Zanubrutinib single agent in the setting of R/R MCL was investigated in multiple trials, showing high efficacy and a long duration of response, with a favorable safety profile [30]. In the phase II registry trial, Zanubrutinib single agent achieved an ORR of 83.7%, with 77.9% of CR and a median duration of response not reached, with median PFS of 33 months, in a cohort of R/R MCL patients [30] (Table 1).
On the basis of the high efficacy of BTKi, ibrutinib was investigated in frontline MCL treatment in addition to standard chemoimmunotherapy. In older patients, the addition of ibrutinib to bendamustine–rituximab (BR) was analyzed in the phase III double-blind randomized SHINE trial, where this combination was compared to BR plus placebo [31]. Among 523 patients, the addition of ibrutinib was demonstrated to significantly improve the CR rate and the progression-free survival (PFS) (80.6 months in the BR plus ibrutinib group and 52.9 months in the BR plus placebo group, HR 0.75, p = 0.01), though overall survival (OS) was similar in the two groups.
Multiple other attempts to ameliorate the standard frontline chemoimmunotherapy have been performed. One of the most relevant was the investigation of second-generation BTKi acalabrutinib in association with BR. The combination BR + acalabrutinib in previously untreated MCL was firstly studied in a phase Ib trial (ACE-LY-106) in both naïve and R/R MCL patients [32], where patients received acalabrutinib from cycle 1 until disease progression or treatment discontinuation; bendamustine on days 1 and 2 of each cycle for up to 6 cycles; and rituximab on day 1 of each cycle for 6 cycles, continuing every other cycle from cycle 8 for 12 additional doses in the first-line cohort. No new safety risks were identified, ORR was 94.4% and 85.0% in the frontline and R/R cohorts, respectively, while CR was 77.8% and 70.0%, respectively. At 47.6 months of follow-up, median PFS and OS were not reached in the treatment naïve group. Thus, a multicenter, double-blind, placebo-controlled phase III trial (ECHO trial) has followed, investigating this combination in a first-line setting, and has been recently presented at the last European Hematological Association (EHA) 2024 Meeting [33]. The study enrolled 598 patients (aged 65 or older), demonstrating a significant amelioration of PFS in the experimental arm (median PFS of 66.4 vs. 49.6 months, respectively, hazard ratio [HR] = 0.73; p = 0.0160) and a trend of better OS.
Another trial, which integrated new drugs in chemoimmunotherapy, is the FIL VR-BAC study [34]. In this trial, patients were allocated as low risk or high risk, depending on the tumor morphology (blastoid versus others), Ki67 expression (≥30% versus others), or presence of TP53 mutation and/or deletion. Patients with any of the three risk factors were classified as HR. Patients with low-risk disease were treated with six cycles of R-BAC (rituximab 375 mg/m2 d 1; bendamustine 70 mg/m2 d 1,2; cytarabine 500 mg/m2 d 1,2,3), while high-risk patients received abbreviated induction with 4 R-BAC, followed by consolidation (4 months, 800 mg/d) and maintenance (20 months, 400 mg/d) with venetoclax. This represents the first prospective study that stratified upfront patients with MCL to different treatments according to their risk profile. The results suggested that the addition of venetoclax to R-BAC improves the performance of the induction strategy, especially in high-risk patients, where the 2-year PFS and OS were, respectively, 58% and 66%, pointing to the importance of identifying high-risk patients from initial diagnosis.
However, all these regimens are based on the bendamustine backbone, that, even if it still represents the standard treatment for elderly MCL patients not eligible for high-dose therapy, opens important issues in the era of novel immunotherapies. In fact, bendamustine is well known to deteriorate T-cell compart and to damage the apheresis product for CAR-T manufacturing [35] A recent report clearly demonstrated that patients exposed to bendamustine had significantly lower CD3+ cells at apheresis and achieved lower ORR, shorter PFS and OS, if compared to not exposed patients. In bendamustine-exposed patients, those with recent treatment (within 9 months) had a worse prognosis. Based on this evidence, bendamustine-backbone treatment should be carefully evaluated in patients who could have CAR-T treatment in their near future, and a minimum time of 9 months from the last dose of bendamustine and apheresis would be desirable in order to not affect the prognosis [35].
In the setting of first-line treatment in young fit MCL patients, the international randomized phase III Triangle Trial has been recently published, representing a new milestone that modified the standard of care [36]. Patients with newly diagnosed stage II–IV MCL, aged 18–65 years and suitable for ASCT, were randomly assigned 1:1:1 to control group A (standard of care) or experimental groups A + I (standard of care with the addition of ibrutinib in both the induction and maintenance phases) or I (ibrutinib administered in the same way as in group A + I, but omitting ASCT). The 3-year failure-free survival (FFS) was significantly higher in the group A + I, if compared to the standard arm (88% vs. 72%, HR 0·52, p = 0·0008). The comparison of group A + I versus group I is ongoing to evaluate if there will still be a role for ASCT after the integration of ibrutinib in the first-line standard of care [26]. At the last ASH 2024, preliminary results of the comparison between arm A + I and I were shown, demonstrating no differences in FFS between the two arms. However, a trend towards the superiority of A + I over I has been suggested in very high-risk patients: Ki-67 > 30%, blastoid morphology, or high p53 expression [36]. On the other side, rituximab maintenance confirmed its benefit in terms of PFS in all the three arms [37].
Completely chemo-free regimens are emerging as another important new paradigm in treatment-naïve MCL patients. Three trials have recently been concluded. Firstly, the ENRICH trail, which investigated the combination of rituximab plus ibrutinib vs. chemoimmunotherapy regimens (R-bendamustine or R-CHOP) in previously untreated over-60-year-old MCL patients [38]. The results of this study were the first demonstrating an improved PFS for rituximab-ibrutinib (RI) versus immunochemotherapy in previously untreated MCL, and this goal was primarily driven by the improved PFS for RI versus RCHOP, while the PFS for IR versus BR was broadly similar. Subgroup analysis suggested a better performance of RI in the low-risk group (such as age < 70, ki67 < 30%, ECOG PS 0, non-blastoid variant, low MIPI score risk). Another important chemo-free experience is the BOVen trial, a phase 2 study of Zanubrutinib, Obinutuzumab, and venetoclax (BOVen) in untreated patients with MCL with a TP53 mutation. In the 25 patients included, the best ORR was 96% (24/25), and the CR was 88% (22/25), with 95% (18/19) of MRD negativity. The 2-year PFS and OS were 72% and 76%, respectively [39]. The third experience is the OAsIs trial (NCT02558816), as shown in Table 2, a single-arm multicenter prospective phase 1/2 trial that aimed to determine the maximum tolerated dose (MTD) of venetoclax in combination with fixed doses of ibrutinib and Obinutuzumab in relapsed MCL patients, with a cohort extension to untreated patients at the venetoclax MTD. Preliminary results showed a CR of 86.6% in untreated patients [40].
MCL is considered an incurable disease. Concerning the R/R MCL setting, huge efforts have been made in the last decade to address this urgent unmet clinical need. Development of Chimeric Antigen Receptor (CAR) T cells (CAR-T cells) completely changed the treatment landscape: in particular, the construct Brexucabtagene autoleucel (Brexu-cel) has been FDA, EMA, and AIFA approved as a third-line treatment for relapsed/refractory MCL, based on the results of the ZUMA-2 study [49,50], which showed that treatment with Brexu-cel induced a durable ORR of 91% and a median duration of response (DOR), PFS, and OS of 28.2 months, 25.8 months, and 46.6 months, respectively, in patients refractory or intolerant to BTKi treatment. These data were confirmed at a longer 3-year follow-up [50] and were demonstrated to be consistent with data coming from real-world practice (real-world evidence, RWE) [51]. Another CD19-directed CAR-T-cell product (Lisocabtagene maraleucel, Liso-cel) for R/R MCL has been evaluated in the phase 1 TRANSCEND NHL 001 study [52]. Results from this trial have recently been published, showing in 88 heavily pretreated patients with R/R MCL an ORR of 83.1%, with a CR rate of 72.3%, and a median PFS of 15.3 months, with a low incidence of grade ≥3 cytokine release syndrome (CRS), neurologic events (Immune effector cell-associated neurotoxicity syndrome, ICANS), and infections in a population of patients affected by high-risk, aggressive disease [51]. These favorable results have been confirmed in RWE, even if higher rates of adverse events emerged: CRS showed to be very frequent (>90% cases), but with a low incidence of grade >3 events; ICANS have been observed in 60–65% of cases, with 36% of grade >3 events [53]. Moreover, RWE showed a high rate of late hematological toxicities and infections risk. The non-relapse mortality rate was 10–15%, with infections as the main cause of death in these patients.
Also, an indirect comparison to a historical cohort of patients treated with chemoimmunotherapy showed a significant survival benefit with CAR-T therapy versus the standard of care in patients with R/R MCL after BTKi exposure [54], strongly suggesting that a CAR-T-cell approach should be offered to these patients.
Moreover, there is some evidence that immunomodulatory drugs, such as lenalidomide, could improve the depth and duration of the response to these T-cell engager treatments, and could be considered in this setting [55,56,57,58].
However, although a remarkable improvement in outcome has been achieved in recent years in both the frontline and R/RMCL, patients still experienced relapses, and a plateau in survival curves has not been reached yet. Here, next-generation therapies are emerging, such as new non-covalent BTKi molecules and novel monoclonal antibodies (MABs), which represent a heterogeneous group of agents, including Antibody–Drug Conjugate (ADC) and T-cell-engaging (TCE) products. The impact of these agents is expected to be meaningful, firstly on R/R disease and subsequently in early treatment lines too.
In this work we will analyze treatment options for MCL patients, who failed or are not eligible for CAR-T-cells treatment, with particular attention to drugs still under development.

4. Role of New Drugs in Relapsed/Refractory MCL After CAR-T Cells or Not Eligible for CAR-T-Cell Therapy

4.1. Currently Available Treatments

BTK inhibitors
Pirtobrutinb
The full mechanisms of resistance to BTKi are still unknown and controversial. The main mechanisms involved seem to be different in chronic lymphocytic leukemia (CLL) versus MCL [37]. In MCL, mutations of the cyclin D1 gene or the CDKN2A/MTAP gene could be the most important ones. Also, the onset of the acquired C481S mutation in the BTK enzyme, which prevents the formation of an irreversible covalent bond between the drug and enzyme, reducing the efficacy of BTKi, has been shown [59,60]. In this setting, Pirtobrutinib (LOXO-305), is a first-in-class, next-generation, highly selective, non-covalent, reversible BTKi with activity against C481-mutated BTK MCL and seems to be effective also in patients resistant to first-generation covalent BTKi.
Pirtobrutinib was investigated in a first-in-human pivotal multicenter phase 1/2 trial, which enrolled 13 patients (9 CLL and 4 MCL) with advanced disease (NCT03740529) (Table 3) [61]. Among patients evaluated for a response, pirtobrutinib treatment showed an 87.5% overall response rate, including a partial response (PR) in CLL patients with the C481 BTK mutation. No dose-limiting toxicities or grade 3 adverse events emerged [61].
Pirtobrutinib was then investigated in monotherapy in the setting of R/R MCL, in the BRUIN trial, an open-label, multicenter phase 1/2 study.
The trial included 61 patients with heavily pretreated R/R MCL. The median number of previous lines of therapy was three: 57 (93%) patients had received a previous BTKi, 60 (98%) an anti-CD20 antibody, 15 (25%) ASCT, 3 (5%) an allogeneic transplant, and 3 (5%) CAR-T-cell therapy. The treatment was demonstrated to be safe and manageable. The most common adverse events reported were fatigue, diarrhea, and contusion. Dose interruptions were observed in a minority of cases. Differently from covalent BTKi, with pirtobrutinib treatment, atrial arrhythmias and hemorrhage were rarely observed (two cases of atrial fibrillation, <1%, and only one case of reported grade 3 hemorrhage, a subarachnoid bleed sustained during a bicycle accident). After six months of follow-up, pirtobrutinib, at the dose of 200 mg daily, showed an ORR of 52%, with 25% of CR 25%, in patients with R/R MCL heavily pretreated and already exposed to BTKi. Responses were observed also in patients with a higher-risk variant (ORR 50% in blastoid variant cases) and in those who received previous cellular therapy, including nine (64%) patients with previous ASCT or allogeneic transplant, and 100% of cases with previous CAR-T-cell therapy. The response also was demonstrated to be early and sustained, with a median time to first response of 1.8 months, with a median duration of response of 8.3 months [41].
On this basis, in January 2023, the U.S. Food and Drug Administration (FDA) granted accelerated approval to pirtobrutinib for the treatment of adult patients with R/R MCL after at least two lines of systemic therapy, including a BTKi [62]. A randomized trial, BRUIN MCL-321 (NCT04662255) [63], is currently open to enrollment and evaluates pirtobrutinib vs. an investigator’s choice of BTKi (ibrutinib, acalabrutinib, or Zanubrutinib) in patients with MCL who have received at least one prior line of therapy and are BTKi-naïve. The primary endpoint is PFS, and secondary endpoints include OS.
  • Ibrutinib + Venetoclax
  • Trial Sympatico
Venetoclax is an oral BCL-2 inhibitor approved in the USA and Europe for the treatment of patients with CLL and previously untreated acute myeloid leukemia [64]. Venetoclax monotherapy was investigated in a phase 1 study in patients with R/R MCL, showing a 75% ORR and a 21% CR rate [42] (Table 2).
Ibrutinib and Venetoclax have distinct and complementary mechanisms of action and have demonstrated synergistic antitumor activity in preclinical models of MCL, showing significantly increase apoptosis (23%) in combinations when compared to each agent used alone (ibrutinib, 3.8%; venetoclax, 3.0%) [65].
On this encouraging basis, the phase 3 international study SYMPATICO (PCYC-1143-CA, NCT03112174) has been conducted, comprising an open-label Safety Run In (SRI) cohort and a subsequent double-blind randomized period, both including R/R MCL patients. An open-label arm in previously untreated patients is also currently ongoing [66,67] (Table 3).
The results of the primary analysis from the randomized, double-blind, phase 3 part of the SYMPATICO study comparing Ibr + Ven vs. Ibr + placebo (Pbo) in pts with R/R MCL were recently presented at the last 2024 American Society of Hematology (ASH) meeting. A total of 267 pts were enrolled and randomly assigned to receive Ibr + Ven (n = 134) or Ibr + Pbo (n = 133). The median age was 68 years old, with an ECOG PS of 0–1 in 96% of cases; 17% had at least three prior lines of treatment, and 22% were at increased risk for tumor lysis syndrome (TLS). With a median time of the study of 51.2 months, the median PFS was significantly longer with Ibr + Ven vs. Ibr + Pbo (31.9 vs. 22.1 months), with a hazard ratio (HR) of 0.65 (95% CI, 0.47–0.88; p = 0.0052). The 2y PFS was 57% and 45% with Ibr + Ven and Ibr + Pbo, respectively, with a consistent PFS benefit across high-risk subgroups, including those with a blastoid variant or TP53-mutated MCL. Grade ≥3 adverse events occurred in 84% of pts with Ibr + Ven vs. 76% with Ibr + Pbo, the most frequent being neutropenia (31% vs. 11%), pneumonia (13% vs. 11%), thrombocytopenia (13% vs. 8%), anemia (10% vs. 3%), diarrhea (8% vs. 2%), leukopenia (7% vs. 0%), and COVID-19 (5% vs. 1%). These data demonstrated that the Ibr + Ven combination led to a significant improvement in PFS compared with Ibr + Pbo in R/R MCL patients, and to a trend of improvement in OS, even if not statistically significant at this interim analysis. The safety profile of Ibr + Ven was consistent with known AEs for each agent, with no new safety signals observed, with an overall favorable benefit–risk profile for this combination in patients with R/R MCL [67,68].

4.2. Treatment Under Investigation but Not Currently Available

A plethora of new targets are being evaluated for MCL (as summarized in Figure 1).
  • Novel Antibodies
  • Antibody–drug conjugate
  • Zilovertamab Vedotin
Zilovertamab vedotin is an immunoconjugate targeting ROR1, carrying the toxin monomethyl auristatin E [44,69,70] (Table 2).
In the first phase 1, first-in-human, dose-escalation study [70], 32 patients with previously treated lymphoid cancers were enrolled to receive Zilovertamab vedotin every 3 weeks until the occurrence of cancer progression or unacceptable toxicity had occurred. The 32 patients presented different histologies (MCL, CLL, diffuse large B-cell lymphoma (DLBCL), FL, Richter transformation lymphoma, or marginal zone lymphoma) and were heavily pretreated (median of four previous treatments). As expected with a monomethyl auristatin E-containing antibody–drug conjugate, adverse events included acute neutropenia and cumulative neuropathy, resulting in a recommended Zilovertamab vedotin dosing regimen of 2.5 mg/kg every 3 weeks. No clinically concerning adverse events occurred to suggest ROR1-mediated toxicities or non-specific binding to normal tissues.
Zilovertamab Vedotin induced objective tumor responses in 7 of 15 patients with MCL (47%; 4 partial and 3 complete) and in 3 of 5 patients with DLBCL (60%; 1 partial and 2 complete); objective tumor responses were not observed among patients with other tumor types.
At the last 2024 ASH, Glimelius et al. [44] presented the results of Cohort A of a phase II, open-label, multicohort, waveLINE-006 study (NCT05458297) of 40 relapsed/refractory MCL patients treated with Zilovertamab Vedotin. The median age was 68 years (range, 42–86), 28 pts (70%) were male, 19 (48%) had an ECOG PS of 0 or 1, 16 (40%) had a high MIPI risk, 7 (18%) were positive for the TP53 mutation, 22 (55%) had a KI67 proliferation fraction ≥30%, and the median number of prior lines of therapy was 4 (range, 2–9). All patients received a covalent or non-covalent BTKi. Eleven pts (28%) had undergone prior autologous stem cell transplant, and six (15%) received prior CAR-T-cell therapy. At the data cutoff, 37 pts (93%) had discontinued treatment (17 PD, 8 adverse even, 5 patient withdrawal, 4 clinical progression, 2 physician decision, 1 non-study anticancer therapy). The ORR was 40%: 5 pts (13%) had a CR and 11 (28%) achieved a PR. The median DOR was 3.0 months, with two responders with a response duration ≥ 6 months. The median PFS was 3.4 months, with a 6-month PFS of 26%. The median OS was 9.0 months, with a 6-month OS of 67%. Treatment-related AEs occurred in 36 pts (90%), of which the most common (≥25%) were neutropenia (58%), peripheral neuropathy (43%; due to conjugated monomethyl auristatine denned as peripheral neuropathy, paresthesia, peripheral sensory neuropathy, and polyneuropathy), and diarrhea (28%). Grade 3 or 4 treatment-related AEs occurred in 32 pts (80%), most commonly neutropenia and peripheral neuropathy. No pts died due to treatment-related AEs. These data confirmed that this setting of patients remains an unmet clinical need; however, the drug-conjugated antibody Zilovertamab vedotin showed promising results and a safe profile in the RR MCL setting, and further investigations are warranted.
  • Loncastuximab-tesirine
Loncastuximab-tesirine is another promising drug-conjugated monoclonal antibody, targeting CD19, conjugated with a pyrrolobenzodiazepine dimer [71,72]. This product was approved by the FDA in April 2021, and subsequently by EMA and AIFA, for the treatment of R/R large B-cell lymphoma (including DLBCL arising from low-grade lymphoma) in adult patients after two or more lines of immune chemotherapy, based on the LOTIS-2 trial [73]. In MCL, loncastuximab-tesirine, administered after a short course of chemotherapy, is under investigation in the COLUMN trial by Fondazione Italiana Linfomi (FIL), currently ongoing (NCT05249959) [74] (Table 2).
Another trial, investigating the safety and efficacy of the combination Loncastuximab Tesirine plus Ibrutinib in DLBCL and MCL (NCT03684694) [75], has recently concluded enrollment, and the results are expected in the near future.
Loncastuximab-tesirine has also been investigated in association with Durvalumab, a human monoclonal Ab of the immunoglobulin G-1 kappa subclass that blocks the interaction of programmed death-ligand 1 (PD-L1) with PD-1 on T cells and with CD80 (B7.1) on other immune cells, in a phase I/II trial (NCT03685344, [76]). Preclinical data, as well as early results from clinical trials combining ADC and checkpoint inhibitors, show the potentially increased effectiveness and synergism of these therapeutics when used in combination. The trial has recently concluded, and the results are expected soon.
Blockade of PD-L1/PD-1 and PD-L1/CD80 interactions releases the inhibition of immune responses, including those that may result in tumor elimination, and provides the rationale for the current trial.
Another ongoing trial is investigating the efficacy of Loncastuximab-tesirine in monotherapy in R/R B-cell malignancies, including MCL (NCT05453396) [48] (Table 2).
However, it is important to notice that Loncastuximab-tesirine treatment is burdened by important adverse events, which can limit its administration. Among them, we find gamma-glutamyltransferase increased (35.8%), neutropenia (34.9%), fatigue (30.2%), anemia (28.8%), thrombocytopenia (28.4%), nausea (26.5%), peripheral oedema (23.3%), and rash (20.0%) [48].
  • Bispecific Antibodies
  • Glofitamab
Glofitamab is a CD20 × CD3 bispecific antibody with an innovative 2:1 tumor–T-cell binding configuration, which translates into bivalent binding to CD20 on B cells and monovalent binding to CD3 on T cells, leading to T-cell engagement and redirection against malignant B cells [77]. Glofitamab is currently approved for the treatment of patients with R/R diffuse large B-cell lymphoma (DLBCL) after >2 lines of therapies [43,78].
Thus, Glofitamab was also investigated in the setting of MCL in the pilot phase I/II NP30179 study (NCT03075696) [79] (Table 2). In this trial, 61 MCL pts were enrolled and received Glofitamab with the standard ramp-up dose, premedicated with Obinutuzumab administered 7 days before the first glofitamab dose (single (1000 mg) or split over 2 days (2000 m) dose). Glofitamab step-up dosing was administered once a day on days 8 (2.5 mg) and 15 (10 mg) of cycle 1, with a target dose of 16 or 30 mg once every 3 weeks from cycle 2 day 1 onward, for 12 cycles. The 2000 mg dose of Obinutuzumab demonstrated reduction of the CRS rate and severity if compared to the single 1000 mg dose. Patients enrolled in the study were heavily pretreated, with a median of two previous therapies (range, 1–5): 51.7% had previously received a BTKi (58.1% of whom received it as their last previous therapy), 73.3% were refractory to their last line of previous therapy, and 36.7% had received previous treatment with bendamustine (22 patients, of whom 9 within the previous 12 months and 4 within the previous 6). With a median follow-up of 19.6 months, the treatment demonstrated high efficacy, with CR and ORR rates of 78.3% (65.8–87.9%) and 85.0% (73.4–92.9%), respectively. Responses were early and durable; the median time to the first response was 42 days, and the median duration of CR (DoCR) was 15.4 months (12.7-NE), with a 12-month DoCR rate of 71.0% (56.8–85.2). The median PFS and OS were 16.8 and 29.9 months, respectively. The patients who were BTKi-naïve showed significantly better outcomes if compared to BTK cases.
Likewise, responses were lower in patients who had received previous bendamustine. The CR and ORR rates were 68.2% (45.1–86.1) and 77.3% (54.6–92.2), respectively; however, these responses were still achieved even when bendamustine had been administered within the previous 12 months (7/9), even after CAR-T administration.
Toxicities were manageable and were most commonly CRS (70.0%), neutropenia (38.3%), COVID-19 (31.7%), and pyrexia (31.7%). Grade 3/4 AEs occurred in 65.0% of patients, most commonly neutropenia (23.3%), pneumonia (11.7%), anemia (11.7%), and CRS (11.7%); the CRS rate was lower in the cohort of patients receiving Obinutuzumab pretreatment 2000 mg vs. 1000 mg (63.6% vs. 87.5%), especially G > 3 events (6.8% vs. 25%). The median time to the CRS onset was 9.7 h and lasted for a median of 49 h after the first glofitamab dose (C1D8-C1D14) and 20.6 or 24.6 h after the second dose (C1D15-C1D21). CRS cases were well managed with the standard tocilizumab and corticosteroid therapy protocols and were especially needed in the Obinutuzumab 1000 mg group, as well as ICU admission (9.1% in the 1000 mg vs. 31.3% in the 2000 mg cohort).
ICANS presented in 11.7% (7/60) of patients and were all G1-G2 AEs and resolved easily.
Glofitamab was interrupted in 60% of patients due to AEs, mainly neutropenia (15%), CRS (10%), infections (COVID-19/COVID pneumonia) (15%), pneumonia 10%, and other infections (6.7%). Eight deaths were reported, all due to infections, though it is important to notice that six out of the eight deaths were COVID-19 related (COVID-19/COVID-19 pneumonia and one post-acute COVID-19 syndrome) due to the trial accrual during the pandemic.
Data on the Glofitamab efficacy in R/R MCL are encouraging; however, follow-up is still limited compared to, for example, with CAR-T, and more mature data are needed, above all on the duration of complete remissions; CD20 expression in this setting of heavily pretreated cases; and the best management of toxicities, such as CRS, though the impact of Obinutuzumab pretreatment on low levels of CRS should be weighted when comparing with other agents. Moreover, glofitamab efficacy needs to be evaluated in all subsets of patients, since a phase 1 pharmacodynamic analysis revealed that increased TP53 gene expression was associated with resistance to glofitamab, conveying a diminished effector T-cell profile [80].
Currently, the GLOBRYTE study is an ongoing phase III, open-label, multi-center, randomized, controlled trial evaluating the efficacy and safety of glofitamab monotherapy in patients with R/RMCL. This study compares glofitamab with the best investigator’s choice of rituximab + bendamustine (BR) or rituximab+ lenalidomide (R-Len) [81]. Results are expected in the near future.
  • Epcoritamab
Epcoritamab is another full-length IgG1 bispecific antibody redirecting CD3 + T cells to CD20-expressing cells [82,83,84]. Many data demonstrated Epcoritamab efficacy in monotherapy and in combinations in relapse/refractory DLBCL and follicular lymphoma. Epcoritamab is already approved in many countries for the treatment of R/R DLBCL after at least two previous lines of therapy. However, data on MCL are still limited: in the dose-escalation part of the pilot study in 73 patients with R/R NHL, only four MCL cases were included [85]. Thus, larger studies focusing on MCL histology are still needed.
  • Mosunetuzumab single agent
Mosunetuzumab is another humanized IgG1-based CD20 × CD3 BsAb with an altered Fc that does not bind to the complement or Fc gamma receptor. It has a single rituximab-like binding site for CD20 and a single binding site for CD3. Like in other bispecific antibodies, also the mosunetuzumab schedule included a dose-escalation phase to reduce the incidence and severity of CRS. The first application of Mosunetuzumab was treatment of R/R follicular lymphoma (FL), revealing a consistent efficacy. The pilot phase I–II study included patients with different subtypes of B-NHL, categorized into indolent non-Hodgkin lymphoma (iNHL); predominantly FL; and aggressive non-Hodgkin lymphoma (aNHL), predominantly DLBCL. Updated results of the study showed an ORR of 65.7 and 36.4%, with a CR rate of 49.3 and 21.7%, in iNHL and aNHL, respectively [86,87,88,89] (Table 3). The mPFS was 12.2 and 1.4 months for iNHL and aNHL, respectively. The efficacy is higher in the third- vs. later-line settings, while recent data demonstrated similar activity for both the disease progression within 24 months after frontline therapy (POD24) and not POD24. Based on early results from this study, mosunetuzumab received FDA and EMA approval in 2022 for the treatment of R/R FL after at least two prior lines of systemic therapy [90].
Mosunetuzumab single agent was also investigated in other settings, such as DLBCL [91,92], marginal zone lymphoma [93], or Richter Syndrome [94].
In MCL, mosunetuzumab monotherapy was studied in an expansion cohort in patients who are R/R to BTKi therapy, administered with the classical step-up dose [45] (Table 2). Twenty-five patients were enrolled, with a median age of 70 years (range: 50–89), Ann Arbor stage III/IV disease in 92%, and a MIPI score ≥6 in 84% of cases. The median number of prior therapies was 3 (range: 2–6), with one-third of cases having received previous ASCT, and 92% were refractory to their most recent regimen. At a median follow-up of 54.5 months, the best ORR and CR rates were 44% and 24%, respectively. Responses were demonstrated to be durable, with a median duration of response and of CR of 10.3 months and 18.0 months, respectively. The median PFS and OS were 3.7 and 7.3 months, respectively. The treatment showed a manageable safety profile, with the most common AEs CRS (52%, predominantly low grade and early), fatigue (36%), and pyrexia (36%). Suspected immune effector cell-associated neurotoxicity syndrome (ICANS) events occurred only in 3 pts (Grade 1 confusional state [n = 2]; Grade 2 delirium [n = 1]), two events concurrently with CRS. Grade 3/4 AEs were reported in 19 (76%) cases, including neutropenia (28%; no febrile neutropenia), hypophosphatemia (20%), thrombocytopenia (16%), and anemia (12%). Serious AEs (excluding fatal PD) were observed in fourteen (56%) pts; ten (40%) were treatment related. No grade 5 fatal AEs were reported. These data suggested mosunetuzumab single-agent activity in a challenging-to-treat population of pts with high-risk and refractory MCL. The safety profile was demonstrated to be manageable, and further investigations are ongoing.
  • Mosunetuzumab combinations
A Phase Ib/II study (NCT03671018), investigating Mosunetuzumab in combination with Polatuzumab-Vedotin (M-Pola) in patients with R/R B cell non-Hodgkin lymphoma, is ongoing (Table 3). Recently, the preliminary safety and efficacy analysis from the ongoing phase II expansion cohort of R/R MCL patients, who had received prior BTKi therapy, were presented [46,95]. Included patients had received ≥2 prior regimens (including an anti-CD20 agent, BTKi, and anthracycline- or bendamustine-based therapy). Mosunetuzumab was administered subcutaneously by step-up dosing on Days 1 (5 mg), 8 (45 mg), and 15 (45 mg) of Cycle 1, then 45 mg on D1 of every cycle from C2D1, every 3 weeks for a total of 17 cycles. Polatuzumab (1.8 mg/kg intravenous infusion) was administered on D1 of the first six courses. At this preliminary analysis, 20 patients had received M-Pola combination treatment. The median age was 68.0 (range 44–82) years, 95% had Ann Arbor stage III/IV disease, and 40% had a MIPI score ≥6 at study entry. The median number of prior lines of therapy was 3 (range 2–9). All cases had received prior BTKi therapy, 7 (35%) had received prior CAR-T-cell therapy, and 85% were refractory to their last therapy. Patients presented high-risk features, with 65% of cases with a Ki-67 proliferation index ≥50%, 50% with blastoid/pleomorphic variants, and 20% with demonstrated TP53 mutation. The ORR and CR rates were 75% and 70%, respectively, with the median duration of CR not yet evaluable. The most common AEs were CRS (50%), fatigue (45%), dyspnea (35%), paresthesia (30%), diarrhea (30%), myalgia (30%), infusion-related reaction (25%), and nausea (25%). Two (10%) toxic deaths occurred, both due to COVID-19 pneumonia. Cases of neuropathy, all grade 1, occurred in three patients (15%). These data are encouraging about this treatment combination in heavily pretreated and very high-risk MCL patients.
  • Odronextamab
Odronextamab is another investigational off-the-shelf CD20 × CD3 bispecific antibody that demonstrated encouraging clinical activity in patients with heavily pretreated R/R diffuse large B-cell lymphoma (DLBCL) in the Phase 2, open-label, multicenter ELM-2 study (NCT03888105;) [96,97] (Table 2).
Thus, Odronexatamab was also investigated in the R/R MCL setting. The Phase 1 ELM-1 study (NCT02290951 [47]) was a single-arm, multicenter, phase 1, dose-escalation and dose-expansion trial, conducted at 10 academic sites across the USA and Germany. A total of 145 heavily pretreated NHL patients were enrolled in the trial, including 85 DLBCL, 40 follicular lymphoma (FL), 12 MCL, 6 marginal zone lymphoma, and 2 other NHL subtypes. In the cohort of 12 R/R MCL, Odronextamab demonstrated encouraging clinical activity, with an ORR of 50%. Patients with MCL showed a higher risk of cytokine release syndrome (CRS) when receiving T-cell engagers due to their circulating tumor burden and other high-risk features: this is reflected by the fact that in 12 patients receiving the initial 1/20 mg step-up regimen in ELM-1, 1 patient (1%) had a Grade 4 CRS event in the context of Grade 5 tumor lysis syndrome, and the global incidence of CRS was higher than in other histologies.
However, since its encouraging efficacy, also in a cohort of R/R MCL with a poor prognosis, that still remained an unmet clinical need; odronextamab is currently under investigation in this setting in the specific cohort of the phase II ELM-2 trial [96]. ELM-2 (NCT03888105) is an ongoing study of odronextamab in patients with R/R B-cell NHL that comprises five disease-specific cohorts, each with independent parallel enrollment: DLBCL, follicular lymphoma, MCL after BTKi therapy, marginal zone lymphoma, and other B-NHL. Patients are being recruited at sites across North America, Europe, and the Asia-Pacific regions. Eligibility criteria for the MCL cohort include age ≥18 years and R/R to ≥1 prior line of systemic therapy, including a BTKi. The MCL cohort will include 78 patients who will receive intravenous odronextamab monotherapy until disease progression or unacceptable toxicity. Following the high rates of observed Grade ≥3 CRS in MCL, including a fatal event in the ELM-1 trial, the step-up regimen was revised to 0.7/4/20 mg to help further mitigate the risk of CRS. At the last updated data presented, as of July 2024, 14 patients with MCL have been enrolled. Results from this trial are expected soon. A sub-analysis of this trial demonstrated that NHLs with greater programmed cell death-ligand 1 expression had a better response to odronextamab therapy; however, this work included only one patient with MCL, so the impact in this disease has not been established yet [98].
  • Allogeneic stem cell transplantation
Allogeneic stem cell transplantation (allo-SCT) can be considered as a post-CAR-T consolidation strategy in patients with relapsed/refractory (R/R) MCL, although prospective evidence is limited [99]. Previous retrospective studies have suggested that allo-SCT may offer a graft-versus-lymphoma (GVL) effect, potentially leading to more durable remissions. In a Spanish retrospective series of 135 patients undergoing allo-SCT, a 5-year event-free survival (EFS) of 47% and an OS of 50% were reported [100]. In this study, the incidence of chronic graft-versus-host disease (cGVHD) was significantly higher in patients who achieved a CR post-transplant (43%) compared to those who did not (0%). The results also demonstrated that chemo-refractoriness is not a significant risk factor for disease control after allo-SCT. The relapse rate was low (7% and 12% at 1 and 3 years, respectively), but the high non-relapse mortality (NRM) may have reduced the number of patients at risk of relapse, since the cumulative incidence of day 100 and one-year NRM were 17% and 32%, respectively. The median age of the patients was 55 years, and the prior lines of therapy were 1–2 in 55% of cases and >2 in 37% of cases. Conditioning was myeloablative in 48% of cases and reduced-intensity conditioning (RIC) in 47% of cases. This series, however, did not include patients with prior exposure to CAR-T-cell therapy; moreover, patients relapsed after CAR-T may be older, with a possible impact on the conditioning regimen, toxicity, and survival.
Di Blasi et al. [101] recently described 238 patients with relapsed/refractory aggressive B-cell lymphomas after CAR-T (both axi-cel and tisa-cel) in France. Relapse/progression was classified as very early (before d +30 days after CAR-T), early (between d +31 and d + 90), and late (>d + 90). The ORR and CR were 14% and 65%, respectively, with the median survival range from 3.7 to 8.5 months. The LDH, ferritin, and CRP levels at infusion and very early relapse were predictive factors for outcomes. To note, no association between previous treatment types and outcomes was observed. However, this cohort lacked MCL patients.
Furthermore, allo-SCT is still complicated by a significant NRM due to infections and GVHD, and can be complicated by the aggressiveness of the disease, poor patient performance status, and/or cytopenias, which can preclude the administration of induction therapy [101]. Liebers et al. [102] compared patients treated with brexucel in the ZUMA-2 study with EBMT MCL patients who underwent allo-SCT by propensity score: at the almost 3-year follow-up, brexucel patients had a significantly higher 1-year OS (81.3% vs. 59.2%, p = 0.004) and lower NRM (3.6% vs. 21.2%, p = 0.015), with cGVHD in 26.9% of alloHCT patients within the first year. However, the long-term progression-free survival and OS remain comparable.
In another work on B-NHL, 16 patients were transplanted after CAR-T failure, with a median PFS and OS of 14.3 and 16.2 months, respectively, from the time of transplant, and the estimated 1-year PFS and OS were 52% and 66%, respectively, with promising results in patients who had achieved a CR at the time of allo-SCT at the 24-month follow-up (6 out of 7 CR still ongoing). However, none of these patients were MCL (13 de novo DLBCL, 2 transformed indolent lymphomas, 6 PMBCL) [103].
Moreover, another experience on allo-SCT in DLBCL reported comparable efficacy, with a 2-year PFS and OS of 31% and 45%, respectively, at the 32-month follow-up but at the cost of considerable toxicity, especially hepatic (28%) and sinusoidal obstruction syndrome (15.4%), and a 2-year non-relapse mortality and relapse/progression incidence of 26% and 43%, respectively [104].
Another encouraging result was reported by Iacoboni et al., where the median OS for 32 patients with R/R DLBCL transplanted after CAR-T was not reached at the 15.1-month follow-up, with a 12-month OS of 84% [105].
Thus, the decision to proceed with allo-SCT after CAR-T-cell therapy must be carefully evaluated, considering the patient’s fitness, the availability of a suitable donor, and the risk of NRM. Further research is needed to establish the optimal criteria for patient selection and the timing of allo-SCT after CAR-T-cell therapy in R/R MCL.
  • Horizon Scanning and Future Perspectives
  • BTK degraders
The continuous treatment with BTKi put selective pressure, leading to multiple BTK mutants. In this setting, a new class of BTK-targeting drugs emerge: the BTK protein degraders, which use the ubiquitin–proteasome pathway to ubiquitinate BTK protein in the cell, leading to proteasomal destruction of BTK. Preclinical experiments show broad activity of BTK protein degraders across the spectrum of wild-type and mutant BTKs, including the most frequent ones identified as mechanisms of resistance in CLL and MCL [106].
The primary results came from the first-in-human drugs NX-2127 and NX-5948.
NX-2127 was investigated in a phase I/Ib trial, with a dose escalation phase and an expansion cohort, enrolling relapsed/refractory B-cell malignancies, including CLL (40 cases, dose 100 mg), MCL (20 patients, dose 300 mg), and DLBCL (20 patients, dose 300 mg) [107] (Table 2).
NX5948 is under investigation in a phase I/Ib trial including CLL, MCL, marginal zone lymphoma, Waldenstrom Disease, follicular lymphoma, DLBCL, and Primary Central Nervous System Lymphoma (Table 2) [108].
Preliminary results of these studies suggested that BTK degraders show early efficacy in clinical trials, with a manageable safety profile that was consistent with previous reports for BTK-targeted therapies, overcoming common resistance mechanisms to kinase inhibitors.
  • Novel Tyrosine Kinase (TK) inhibitors
Narazaciclib (ON123300) is a second-generation, orally bioavailable, and clinical-stage CDK4/6 inhibitor (CDKi) that may trigger cell cycle arrest and significant tumor growth inhibition (TGI) in BTKi-resistant MCL [109]. It was demonstrated to be safe and effective in in vitro and in vivo models of MCL [110].
KIN-8194, a Src-family tyrosine kinase hematopoietic cell kinase (HCK) inhibitor, arrests growth and the integrin-mediated adhesion of BTKi-sensitive MCL cells, as well as MCL cells with primary or acquired BTKi resistance, and faces the therapeutic landscape as a promising novel treatment for MCL patients.
  • Novel BCL-2 inhibitors
Several preclinical studies have suggested BCL-2 as a possible co-target for R/R MCL, and the SYMPATICO study has paved the way for the testing of new BCL-2 inhibitors, often to overcome venetoclax resistance [111,112,113]. The efficacy of obataclax, a BH3 mimetic inhibitor of anti-apoptotic Bcl-2 proteins, which demonstrated synergy with bortezomib in preclinical models, was studied in a phase 1–2 trial in association with bortezomib for 13 R/R MCL patients. The combination was overall well tolerated; however, the ORR was 31% (4/13), and the increased effectiveness compared to bortezomib alone was not confirmed [114].
Another molecule investigated is BGB-11417 (sonrotoclax), a highly selective Bcl-2 inhibitor with 5-fold increased potency in pharmacodynamic studies [115]. Phase I data in patients with R/R MCL showed an ORR of 55% when used in combination with Zanubrutinib (Table 2) [116].
Further studies are needed to confirm these promising results in a larger population.
  • AKT-inhibitors
Capivasertib is a novel oral pan-AKT inhibitor, which has been studied in a phase II trial including 2 MCL patients among the 15 total patients. The authors reported an ORR of 54% and a CR rate of 8%. A grade 3 AE occurred in three patients (20%), without a G ≥ 3 infection, displaying a manageable safety profile in heavily pretreated NHL [117].
  • Novel CAR-T cells
Another promising field of research is investigating novel CAR-T-cells constructs, direct towards new targets.
In this landscape, of particular interest is a new anti-ROR1-specific CAR-T cell (Onct-808), which has been investigated in a phase 1/2 multicenter study in patients with R/R aggressive B-cell lymphoma [118]. At the last European Hematology Association (EHA) meeting, the results on the first four enrolled patients were presented. Of the four treated patients, three cases had R/R MCL, and two patients received prior CD19 CAR-T cells. Toxicities were relevant, with one 80-year-old patient dying from CRS and ICANS complications, and two cases of grade 3 pneumonia, and due to clinical and economic reasons, the trial was terminated. Response assessments were evaluated in three pts, with CR observed at Month 1 for two of three pts, while the third experienced PR.
Another promising product investigated in MCL is LV20.19 CAR, a bispecific, tandem, lentiviral CAR-T cell targeting both CD20 and CD19 B-cell antigens, with 4-1BB co-stimulatory signaling. This peculiar construct will improve an expansion in IL7 and IL15, producing less “exhausted” CAR product, resulting in more durable clinical activity. Shah et al. conducted a phase 1/2 single-center, prospective trial (NCT04186520) evaluating LV20.19 CAR-T cells at a fixed dose of 2.5 × 106 cells/kg for patients with R/R NHL and presented the results of the MCL cohort [119]. Ten MCL patients received LV20.19 CAR-T cells; manufacturing was successful in 100% of cases, and all received fresh (non-cryopreserved) product. The clinical characteristics were median age of 62 years, with a median of 4 (range 3–8) previous lines of therapy. All patients were heavily pretreated: three cases received prior ASCT and two patients allogeneic HCT; 80% had progressed on a BTKi, and four of these patients had additionally progressed on a non-covalent BTKi (pirtobrutinib) administered on a clinical trial. The Day 28 ORR was 100% (CR = 60% and PR = 40%). Seven of nine pts with MRD assessed between Day 28 and 60 post-CAR were negative. No patient has relapsed, with a median follow-up of 18 months among surviving pts. Regarding the toxicity profile, 100% of patients experienced a Grade 1–2 CRS (but no grade >3 events were observed). ICANS occurred in two cases, with one patient experiencing Grade 2 and the other Grade 3 toxicity. At 1 year, the cumulative incidence of relapse and NRM were 0% and 10% (one patient died because of gram-negative sepsis after the Day 28 evaluation), respectively, while the 1-year PFS and OS were 90% each. These data suggested that dual targeting of CD19 and CD20 with CAR-T cells may improve outcomes in R/R MCL patients; but data on a larger cohort and longer follow-up are needed [120].
Many other products are on the horizon, scanning for R/R MCL, such as Milatuzumab, a fully humanized anti-CD74 monoclonal antibody, which demonstrated significant activity in preclinical lymphoma models but failed to provide meaningful benefits in clinical trials, mainly due to its short half-life. This could be overcome targeting CD74 using a CAR-T cell; that would probably provide potent and durable anti-MCL activity [121]. Further studies in this direction are ongoing.
  • Exploratory trials
Several new targets are being investigated in a number of active phase I–II trials, both as single agents and in combination with other drugs, both already in use and experimental, as shown in Table 2.
They include, in particular, copanlisib (PI3K inhibitor, NCT04939272), linperlisib (PI3Kδ inhibitor, NCT06324994), alisertib (selective aurora A kinase inhibitor, NCT01695941), avelumab (anti PD-L1 MAB) and utomilumab (anti-human 4-1BB MAB, NCT03440567), nemtabrutinib (novel BTKi, NCT03162536), rocbrutinib (novel BTKi, NCT05716087 and NCT04775745), CYT-0851 (MCT-mediated lactate transport inhibitor, NCT03997968), NX-2127 (BTK degrader, NCT04830137) and NX5948 (BTK degrader, NCT05131022), TQB3909 (BCL-2 inhibitor, NCT06106841), novel anti-CD19 CAR-T (NCT04484012), anti-ROR1 CAR-T (NCT05444322), CLIC-2201 (anti-CD22 CAR-T, NCT06208735), NVG-111 (ROR1-CD3 bispecific antibody, NCT04763083), anti-BAFFR CAR-T (NCT05370430), CD19 CAR NK/T (NCT06464861), SynKIR-310 (anti-CD19 KIR CAR-T, NCT06544265), SC262 (anti-CD22 CAR-T, NCT06285422), GLPG5101 (anti-CD19 CAR-T, NCT06561425), ATA3219 (allogeneic anti-CD19 CAR-T, NCT06256484), CAR.k.28 (NCT04223765), iopofosine (phospholipid drug conjugate delivering I-131, NCT02952508), pemigatinib (FGFR2 inhibitor, NCT06300528), CTX112 (allogeneic CRISPR-Cas9 engineered anti-CD19 CAR-T, NCT05643742), voruciclib (CDK9 inhibitor, NCT03547115), AC676 (BTK degrader, NCT05780034), AZD5492 (CD8-Guided T-Cell Engager, NCT06542250), and PRT2527 (CDK9 inhibitor, NCT05665530); and already mentioned drugs like pirtobrutinib, in addition to glofitamab (NCT06252675), BGB-11417 (novel BCL-2 inhibitor, NCT05471843), LV20.19 CAR-T (NCT04186520), loncastuximab (anti-CD19 ICD, NCT05249959 and NCT05453396), and odronextamab (NCT03888105).
Some trials also use already known drugs, such as carfilzomib and ixazomib (proteasome inhibitors), lenalidomide (immunomodulatory drugs), polatuzumab vedotin (anti-CD79b ICD), tafasitamab (anti-CD19 MAB), nivolumab (anti-PD-1 MAB), oral azacitidine (demethylating agent), anti-CD20 CAR-T (NCT03277729), blinatumomab (anti-CD19 MAB), and Zanubrutinib (BTKi). The first results of these pioneering trials are eagerly awaited.

5. Conclusions

In recent years, remarkable progress in the immunotherapy of hematologic malignancies has significantly improved outcomes for patients with R/R lymphoid diseases. In mantle cell lymphoma, the anticipation of BTK inhibitors in the first-line setting and the spreading of CAR-T-cells therapy completely changed the landscape. However, many cases do not yet respond or relapse after CAR-T cells, and a subset of patients are not eligible for CAR-T in the first place. This setting of MCL patients still represents a challenge and an unmet clinical need. Next-generation therapies are emerging, such as new non-covalent BTKi molecules and novel monoclonal antibodies (MABs), which represent a heterogeneous group of agents, including Antibody drug-conjugated (ADC) and T-cell-engaging (TCE) products. The impact of these agents is expected to be meaningful, firstly on relapsed/refractory (R/R) disease and subsequently also in earlier treatment lines.

6. Future Directions

The near future in mantle cell lymphoma will see the employment of BTK inhibitors in frontline treatment. Also, biological high-risk features, such as TP53 mutations or the ki67 proliferation index, will help, maybe more than age and fitness, to guide clinicians in the treatment choice. The landscape of R/R patients who failed or were ineligible for CAR-T cells still remains a challenge, and novel monoclonal antibodies and novel combinations will probably play a key role in this setting.
In this scenario (Figure 2), future treatment sequencing for young and/or fit patients may be considered as chemoimmunotherapy plus ibrutinib frontline; at the first relapse, CAR-T cells are not allowed in the present moment but may be anticipated in the close future. Actually, in the second line, we can consider BTKi again; eventually non-covalent BTKi, such as pirtobrutinib; followed by CAR-T cells at the second relapse. In elderly patients, first-line treatment is still mainly represented by chemoimmunotherapy with bendamustine-containing regimens, with eventual addition of BTKi (ibrutinib or acalabrutinib), which may impact the later efficacy of TCE, especially in early relapse at less than 9 months from the last bendamustine dose. In the near future, we may have chemo-free regimens available. The second-line therapy can be still represented by BTKi. In patients who fail CAR-T cells, or who are not eligible for that kind of intensive treatment, bispecific antibodies, even if at the present moment not available yet, will probably achieve a key role in the future, especially in early CD20-positive relapses, followed by an ADC like zilovertamab-vedotin or loncastuximab as a last resort. Novel therapies, such as BTK-degraders or novel BCL2 inhibitors and novel combinations, still need to be deeper investigated but will probably have a growing role in the close future in R/R MCL patients, moreover, with progressive anticipation of other drugs in earlier therapeutic lines.

Author Contributions

Conceptualization, A.C., E.B. and L.C.; methodology, A.C., E.B. and L.C.; software, L.C.; validation, A.C., E.B. and L.C.; formal analysis, not applicable; investigation, not applicable; resources, not applicable; data curation, not applicable; writing—original draft preparation, E.B., L.C., R.T. and A.C.; writing—review and editing, A.C., R.T., C.C., M.F., M.L., M.M. and D.M.; visualization, A.C., E.B. and L.C.; supervision, A.C.; project administration, A.C., E.B. and L.C.; funding acquisition, D.M. All authors have read and agreed to the published version of the manuscript.

Funding

The APC was funded by A.I.L. di Cuneo—Paolo Rubino—ODV, V. Schiaparelli, 23—12100 CUNEO, CF 80102390582.

Data Availability Statement

No new data were created or analyzed in this study. Data sharing is not applicable to this article. The original contributions presented in this study are included in the article. Further inquiries can be directed to the corresponding author(s). Images created in BioRender. Comba, L (2025). https://biorender.com/edv7htl (accessed on 26 June 2025); https://biorender.com/yv371pa (accessed on 26 June 2025).

Conflicts of Interest

E.B.: honoraria from Astrazeneca, Amgen, Beigene, Mayo Clinic, travel grants from Kyowa Kirin. M.L.: reports consulting fees from Astrazeneca, Beigene, Janssen and Lilly. Honoraria for lectures, speaker’s bureaus or educational events from Astrazeneca, Beigene, Janssen and Lilly. Participation on data safety monitoring board with Acerta. C.A. speaker’s bureau and travel grant from Takeda. Honoraria for lectures from Taked and Novartis. A.C.: travel grant from Kyowa Kirin and Kite, spearker’s bureau from Roche and Kite. R.T.: honoraria from Lilly and Janssen. M.F.: honoraria from Astrazeneca, travel grant from Kite. M.M., L.C. and D.M. declare no conflict of interest.

Abbreviations

MCLmantle cell lymphoma
CAR-T cellschimeric antigen receptor-T cells
BTKiBruton’s tyrosine kinase inhibitor
MABmonoclonal antibody
ICDimmunoconjugated drug
TCET-cell engaging
R/Rrelapsed/refractory
NHLnon-Hodgkin lymphoma
COOcell of origin
MIPIMCL international prognostic index
MRDminimal residual disease
ASCTautologous stem cell transplantation
ORRoverall response rate
CRcomplete remission
PFSprogression-free survival
OSoverall survival
DORduration of response
BRBendamustine-rituximab
BACBendamustine-cytarabine-rituximab
R-CHOPrituximab-prednisone-doxorubicin-vincristine-cyclophosphamide
MTDmaximum tolerated dose
Brexu-celBrexucabtagene autoleucel
Liso-celLisocabtagene maraleucel
CLLchronic lymphocytic leukemia
TLStumor lysis syndrome
CRScytokine release syndrome
ICANSimmune effector cell-associated neurotoxicity syndrome
ROR1retinoic acid receptor 1
PD-L1programmed death-ligand 1
DLBCLdiffuse large B-cell lymphoma
FLfollicular lymphoma

References

  1. Dreyling, M.; Campo, E.; Hermine, O.; Jerkeman, M.; Le Gouill, S.; Rule, S.; Shpilberg, O.; Walewski, J.; Ladetto, M. Newly diagnosed and relapsed mantle cell lymphoma: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up. Ann. Oncol. 2017, 28 (Suppl. 4), iv62–iv71. [Google Scholar] [CrossRef] [PubMed]
  2. Romaguera, J.E.; Medeiros, L.J.; Hagemeister, F.B.; Fayad, L.E.; Rodriguez, M.A.; Pro, B.; Younes, A.; McLaughlin, P.; Goy, A.; Sarris, A.H.; et al. Frequency of gastrointestinal involvement and its clinical significance in mantle cell lymphoma. Cancer 2003, 97, 586–591. [Google Scholar] [CrossRef] [PubMed]
  3. Silkenstedt, E.; Dreyling, M. Mantle cell lymphoma-Update on molecular biology, prognostication and treatment approaches. Hematol. Oncol. 2023, 41 (Suppl. 1), 36–42. [Google Scholar] [CrossRef] [PubMed]
  4. Campo, E.; Jaffe, E.S.; Cook, J.R.; Quintanilla-Martinez, L.; Swerdlow, S.H.; Anderson, K.C.; Brousset, P.; Cerroni, L.; de Leval, L.; Dirnhofer, S.; et al. The International Consensus Classification of Mature Lymphoid Neoplasms: A report from the Clinical Advisory Committee. Blood 2022, 140, 1229–1253. [Google Scholar] [CrossRef]
  5. Puente, X.S.; Jares, P.; Campo, E. Chronic lymphocytic leukemia and mantle cell lymphoma: Crossroads of genetic and microenvironment interactions. Blood 2018, 131, 2283–2296. [Google Scholar] [CrossRef]
  6. Alaggio, R.; Amador, C.; Anagnostopoulos, I.; Attygalle, A.D.; Araujo, I.B.O.; Berti, E.; Bhagat, G.; Borges, A.M.; Boyer, D.; Calaminici, M.; et al. The 5th edition of the World Health Organization Classification of Haematolymphoid Tumours: Lymphoid Neoplasms. Leukemia 2022, 36, 1720–1748. [Google Scholar] [CrossRef]
  7. Hoster, E.; Rosenwald, A.; Berger, F.; Bernd, H.W.; Hartmann, S.; Loddenkemper, C.; Barth, T.F.; Brousse, N.; Pileri, S.; Rymkiewicz, G.; et al. Prognostic Value of Ki-67 Index, Cytology, and Growth Pattern in Mantle-Cell Lymphoma: Results From Randomized Trials of the European Mantle Cell Lymphoma Network. J. Clin. Oncol. 2016, 34, 1386–1394. [Google Scholar] [CrossRef]
  8. Hoster, E.; Dreyling, M.; Klapper, W.; Gisselbrecht, C.; van Hoof, A.; Kluin-Nelemans, H.C.; Pfreundschuh, M.; Reiser, M.; Metzner, B.; Einsele, H.; et al. A new prognostic index (MIPI) for patients with advanced-stage mantle cell lymphoma. Blood 2008, 111, 558–565. [Google Scholar] [CrossRef]
  9. Aukema, S.M.; Hoster, E.; Rosenwald, A.; Canoni, D.; Delfau-Larue, M.H.; Rymkiewicz, G.; Thorns, C.; Hartmann, S.; Kluin-Nelemans, H.; Hermine, O.; et al. Expression of TP53 is associated with the outcome of MCL independent of MIPI and Ki-67 in trials of the European MCL Network. Blood 2018, 131, 417–420. [Google Scholar] [CrossRef]
  10. Eskelund, C.W.; Dahl, C.; Hansen, J.W.; Westman, M.; Kolstad, A.; Pedersen, L.B.; Montano-Almendras, C.P.; Husby, S.; Freiburghaus, C.; Ek, S.; et al. TP53 mutations identify younger mantle cell lymphoma patients who do not benefit from intensive chemoimmunotherapy. Blood 2017, 130, 1903–1910. [Google Scholar] [CrossRef]
  11. Hermine, O.; Jiang, L.; Walewski, J.; Bosly, A.; Thieblemont, C.; Szymczyk, M.; Pott, C.; Salles, G.; Feugier, P.; Hübel, K.; et al. High-Dose Cytarabine and Autologous Stem-Cell Transplantation in Mantle Cell Lymphoma: Long-Term Follow-Up of the Randomized Mantle Cell Lymphoma Younger Trial of the European Mantle Cell Lymphoma Network. J. Clin. Oncol. 2023, 41, 479–484. [Google Scholar] [CrossRef] [PubMed]
  12. Pott, C.; Hoster, E.; Delfau-Larue, M.H.; Beldjord, K.; Böttcher, S.; Asnafi, V.; Plonquet, A.; Siebert, R.; Callet-Bauchu, E.; Andersen, N.; et al. Molecular remission is an independent predictor of clinical outcome in patients with mantle cell lymphoma after combined immunochemotherapy: A European MCL intergroup study. Blood 2010, 115, 3215–3223. [Google Scholar] [CrossRef] [PubMed]
  13. Yi, S.; Yan, Y.; Jin, M.; Bhattacharya, S.; Wang, Y.; Wu, Y.; Yang, L.; Gine, E.; Clot, G.; Chen, L.; et al. Genomic and transcriptomic profiling reveals distinct molecular subsets associated with outcomes in mantle cell lymphoma. J. Clin. Invest. 2022, 132, e153283. [Google Scholar] [CrossRef] [PubMed]
  14. Tang, G.; Jain, P.; Hu, S.; Ok, C.Y.; Wang, W.J.; Quesada, A.E.; Wei, Q.; Li, S.; Xu, J.; Loghavi, S.; et al. Optical genome mapping reveals diverse mechanisms of cyclin activation in mantle cell lymphomas lacking IGH::CCND1. Hum. Pathol. 2025, 159, 105793. [Google Scholar] [CrossRef]
  15. Lazarian, G.; Chemali, L.; Bensalah, M.; Zindel, C.; Lefebvre, V.; Thieblemont, C.; Martin, A.; Tueur, G.; Letestu, R.; Fleury, C.; et al. TP53 Mutations Detected by NGS Are a Major Clinical Risk Factor for Stratifying Mantle Cell Lymphoma. Am. J. Hematol. 2025, 100, 933–936. [Google Scholar] [CrossRef]
  16. Zoellner, A.K.; Unterhalt, M.; Stilgenbauer, S.; Hübel, K.; Thieblemont, C.; Metzner, B.; Topp, M.; Truemper, L.; Schmidt, C.; Bouabdallah, K.; et al. Long-term survival of patients with mantle cell lymphoma after autologous haematopoietic stem-cell transplantation in first remission: A post-hoc analysis of an open-label, multicentre, randomised, phase 3 trial. Lancet Haematol. 2021, 8, e648–e657. [Google Scholar] [CrossRef]
  17. Le Gouill, S.; Thieblemont, C.; Oberic, L.; Moreau, A.; Bouabdallah, K.; Dartigeas, C.; Damaj, G.; Gastinne, T.; Ribrag, V.; Feugier, P.; et al. Rituximab after Autologous Stem-Cell Transplantation in Mantle-Cell Lymphoma. N. Engl. J. Med. 2017, 377, 1250–1260. [Google Scholar] [CrossRef]
  18. Rummel, M.J.; Niederle, N.; Maschmeyer, G.; Banat, G.A.; von Grünhagen, U.; Losem, C.; Kofahl-Krause, D.; Heil, G.; Welslau, M.; Balser, C.; et al. Bendamustine plus rituximab versus CHOP plus rituximab as first-line treatment for patients with indolent and mantle-cell lymphomas: An open-label, multicentre, randomised, phase 3 non-inferiority trial. Lancet 2013, 381, 1203–1210. [Google Scholar] [CrossRef]
  19. Tisi, M.C.; Moia, R.; Patti, C.; Evangelista, A.; Ferrero, S.; Spina, M.; Tani, M.; Botto, B.; Celli, M.; Puccini, B.; et al. Long-term follow-up of rituximab plus bendamustine and cytarabine in older patients with newly diagnosed MCL. Blood Adv. 2023, 7, 3916–3924. [Google Scholar] [CrossRef]
  20. Robak, T.; Jin, J.; Pylypenko, H.; Verhoef, G.; Siritanaratkul, N.; Drach, J.; Raderer, M.; Mayer, J.; Pereira, J.; Tumyan, G.; et al. Frontline bortezomib, rituximab, cyclophosphamide, doxorubicin, and prednisone (VR-CAP) versus rituximab, cyclophosphamide, doxorubicin, vincristine, and prednisone (R-CHOP) in transplantation-ineligible patients with newly diagnosed mantle cell lymphoma: Final overall survival results of a randomised, open-label, phase 3 study. Lancet Oncol. 2018, 19, 1449–1458. [Google Scholar] [CrossRef]
  21. Visco, C.; Di Rocco, A.; Evangelista, A.; Quaglia, F.M.; Tisi, M.C.; Morello, L.; Zilioli, V.R.; Rusconi, C.; Hohaus, S.; Sciarra, R.; et al. Outcomes in first relapsed-refractory younger patients with mantle cell lymphoma: Results from the MANTLE-FIRST study. Leukemia 2021, 35, 787–795. [Google Scholar] [CrossRef] [PubMed]
  22. Wang, M.L.; Rule, S.; Martin, P.; Goy, A.; Auer, R.; Kahl, B.S.; Jurczak, W.; Advani, R.H.; Romaguera, J.E.; Williams, M.E.; et al. Targeting BTK with ibrutinib in relapsed or refractory mantle-cell lymphoma. N. Engl. J. Med. 2013, 369, 507–516. [Google Scholar] [CrossRef] [PubMed]
  23. Dreyling, M.; Jurczak, W.; Jerkeman, M.; Silva, R.S.; Rusconi, C.; Trneny, M.; Offner, F.; Caballero, D.; Joao, C.; Witzens-Harig, 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, 770–778. [Google Scholar] [CrossRef]
  24. Wang, M.; Rule, S.; Zinzani, P.L.; Goy, A.; Casasnovas, O.; Smith, S.D.; Damaj, G.; Doorduijn, J.; Lamy, T.; Morschhauser, F.; et al. Acalabrutinib in relapsed or refractory mantle cell lymphoma (ACE-LY-004): A single-arm, multicentre, phase 2 trial. Lancet 2018, 391, 659–667. [Google Scholar] [CrossRef]
  25. Le Gouill, S.; Długosz-Danecka, M.; Rule, S.; Zinzani, P.L.; Goy, A.; Smith, S.D.; Doorduijn, J.K.; Panizo, C.; Shah, B.D.; Davies, A.J.; et al. Final results and overall survival data from a phase II study of acalabrutinib monotherapy in patients with relapsed/refractory mantle cell lymphoma, including those with poor prognostic factors. Haematologica 2024, 109, 343–350. [Google Scholar] [CrossRef]
  26. Zinzani, P.L.; Mauro, F.R.; Tedeschi, A.; Varettoni, M.; Zaja, F.; Barosi, G. Unmet clinical needs in the use of zanubrutinib in malignant lymphomas (Waldenström macroglobulinemia, marginal zone lymphoma and mantle cell lymphoma): A consensus-based position paper from an ad hoc expert panel. Hematol. Oncol. 2023, 41, 795–808. [Google Scholar] [CrossRef]
  27. Tam, C.S.; Opat, S.; Simpson, D.; Cull, G.; Munoz, J.; Phillips, T.J.; Kim, W.S.; Rule, S.; Atwal, S.K.; Wei, R.; et al. Zanubrutinib for the treatment of relapsed or refractory mantle cell lymphoma. Blood Adv. 2021, 5, 2577–2585. [Google Scholar] [CrossRef]
  28. Song, Y.; Zhou, K.; Zou, D.; Zhou, J.; Hu, J.; Yang, H.; Zhang, H.; Ji, J.; Xu, W.; Jin, J.; et al. Treatment of Patients with Relapsed or Refractory Mantle-Cell Lymphoma with Zanubrutinib, a Selective Inhibitor of Bruton’s Tyrosine Kinase. Clin. Cancer Res. 2020, 26, 4216–4224. [Google Scholar] [CrossRef]
  29. Flinsenberg, T.W.H.; Tromedjo, C.C.; Hu, N.; Liu, Y.; Guo, Y.; Thia, K.Y.T.; Noori, T.; Song, X.; Aw Yeang, H.X.; Tantalo, D.G.; et al. Differential effects of BTK inhibitors ibrutinib and zanubrutinib on NK-cell effector function in patients with mantle cell lymphoma. Haematologica 2020, 105, e76–e79. [Google Scholar] [CrossRef]
  30. Song, Y.; Zhou, K.; Zou, D.; Zhou, J.; Hu, J.; Yang, H.; Zhang, H.; Ji, J.; Xu, W.; Jin, J.; et al. Zanubrutinib in relapsed/refractory mantle cell lymphoma: Long-term efficacy and safety results from a phase 2 study. Blood 2022, 139, 3148–3158. [Google Scholar] [CrossRef]
  31. Wang, M.L.; Jurczak, W.; Jerkeman, M.; Trotman, J.; Zinzani, P.L.; Belada, D.; Boccomini, C.; Flinn, I.W.; Giri, P.; Goy, A.; et al. Ibrutinib plus Bendamustine and Rituximab in Untreated Mantle-Cell Lymphoma. N. Engl. J. Med. 2022, 386, 2482–2494. [Google Scholar] [CrossRef] [PubMed]
  32. Phillips, T.; Wang, M.; Robak, T.; Gallinson, D.; Stevens, D.; Patel, K.; Ramadan, S.; Wun, C.C.; Jurczak, W.; Smith, S.D. Safety and efficacy of acalabrutinib plus bendamustine and rituximab in patients with treatment-naïve or relapsed/refractory mantle cell lymphoma: Phase Ib trial. Haematologica 2025, 110, 715–724. [Google Scholar] [CrossRef] [PubMed]
  33. Dreyling, M.; Mayer, J.; Belada, D.; Song, Y.; Jurczak, W.; Paludo, J.; Chu, M.P.; Kryachok, I.; Fogliatto, L.M.; Cheah, C.Y.; et al. High-Risk Subgroups and MRD: An Updated Analysis of the Phase 3 ECHO Trial of Acalabrutinib with Bendamustine/Rituximab in Previously Untreated Mantle Cell Lymphoma. Blood 2024, 144 (Suppl. 1), 1626. [Google Scholar] [CrossRef]
  34. Visco, C.; Tabanelli, V.; Sacchi, M.V.; Evangelista, A.; Ferrarini, I.; Tisi, M.C.; Merli, A.; Fiori, S.; Zilioli, V.R.; Re, A.; et al. Rituximab, Bendamustine and Cytarabine Followed By Venetoclax (V-RBAC) in High-Risk Older Patients with Mantle Cell Lymphoma: A Phase 2 Study By the Fondazione Italiana Linfomi (FIL). Blood 2023, 142 (Suppl. 1), 737. [Google Scholar] [CrossRef]
  35. Iacoboni, G.; Navarro, V.; Martín-López, A.; Rejeski, K.; Kwon, M.; Jalowiec, K.A.; Amat, P.; Reguera-Ortega, J.L.; Gallur, L.; Blumenberg, V.; et al. Recent Bendamustine Treatment Before Apheresis Has a Negative Impact on Outcomes in Patients With Large B-Cell Lymphoma Receiving Chimeric Antigen Receptor T-Cell Therapy. J. Clin. Oncol. 2024, 42, 205–217. [Google Scholar] [CrossRef]
  36. Dreyling, M.; Doorduijn, J.; Giné, E.; Jerkeman, M.; Walewski, J.; Hutchings, M.; Mey, U.; Riise, J.; Trneny, M.; Vergote, V.; et al. Ibrutinib combined with immunochemotherapy with or without autologous stem-cell transplantation versus immunochemotherapy and autologous stem-cell transplantation in previously untreated patients with mantle cell lymphoma (TRIANGLE): A three-arm, randomised, open-label, phase 3 superiority trial of the European Mantle Cell Lymphoma Network. Lancet 2024, 403, 2293–2306. [Google Scholar] [CrossRef]
  37. Ladetto, M.; Gutmair, K.; Doorduijn, J.K.; Gine, E.; Jerkeman, M.; Walewski, J.; Hutchings, M.; Mey, U.; Riise, J.; Trneny, M.; et al. Impact of Rituximab Maintenance Added to Ibrutinib-Containing Regimens with and without ASCT in Younger, Previously Untreated MCL Patients: An Analysis of the Triangle Data Embedded in the Multiply Project. Blood 2024, 144 (Suppl. 1), 237. [Google Scholar] [CrossRef]
  38. Lewis, D.J.; Jerkeman, M.; Sorrell, L.; Wright, D.; Glimelius, I.; Pasanen, A.; Christensen, J.H.; Wader, K.F.; Davies, A.J.; Morley, N.; et al. Ibrutinib-Rituximab Is Superior to Rituximab-Chemotherapy in Previously Untreated Older Mantle Cell Lymphoma Patients: Results from the International Randomised Controlled Trial, Enrich. Blood 2024, 144 (Suppl. 1), 235. [Google Scholar] [CrossRef]
  39. Kumar, A.; Soumerai, J.; Abramson, J.S.; Barnes, J.A.; Caron, P.; Chhabra, S.; Chabowska, M.; Dogan, A.; Falchi, L.; Grieve, C.; et al. Zanubrutinib, obinutuzumab, and venetoclax for first-line treatment of mantle cell lymphoma with a TP53 mutation. Blood 2025, 145, 497–507. [Google Scholar] [CrossRef]
  40. Le Gouill, S.; Morschhauser, F.; Chiron, D.; Bouabdallah, K.; Cartron, G.; Casasnovas, O.; Bodet-Milin, C.; Ragot, S.; Bossard, C.; Nadal, N.; et al. Ibrutinib, obinutuzumab, and venetoclax in relapsed and untreated patients with mantle cell lymphoma: A phase 1/2 trial. Blood 2021, 137, 877–887. [Google Scholar] [CrossRef]
  41. Mato, A.R.; Shah, N.N.; Jurczak, W.; Cheah, C.Y.; Pagel, J.M.; Woyach, J.A.; Fakhri, B.; Eyre, T.A.; Lamanna, N.; Patel, M.R.; et al. Pirtobrutinib in relapsed or refractory B-cell malignancies (BRUIN): A phase 1/2 study. Lancet 2021, 397, 892–901. [Google Scholar] [CrossRef] [PubMed]
  42. Davids, M.S.; Roberts, A.W.; Seymour, J.F.; Pagel, J.M.; Kahl, B.S.; Wierda, W.G.; Puvvada, S.; Kipps, T.J.; Anderson, M.A.; Salem, A.H.; et al. Phase I First-in-Human Study of Venetoclax in Patients With Relapsed or Refractory Non-Hodgkin Lymphoma. J. Clin. Oncol. 2017, 35, 826–833. [Google Scholar] [CrossRef] [PubMed]
  43. Hutchings, M.; Morschhauser, F.; Iacoboni, G.; Carlo-Stella, C.; Offner, F.C.; Sureda, A.; Salles, G.; Martínez-Lopez, J.; Crump, M.; Thomas, D.N.; et al. Glofitamab, a Novel, Bivalent CD20-Targeting T-Cell-Engaging Bispecific Antibody, Induces Durable Complete Remissions in Relapsed or Refractory B-Cell Lymphoma: A Phase I Trial. J. Clin. Oncol. 2021, 39, 1959–1970. [Google Scholar] [CrossRef] [PubMed]
  44. Glimelius, I.; Kim, W.S.; Paszkiewicz-Kozik, E.; Ernst, D.; Merryman, R.W.; Moreira, C.; Tu, S.; Ren, Y.; Ryland, K.; Ogbu, U.C.; et al. Zilovertamab Vedotin Monotherapy for Patients with Relapsed or Refractory Mantle Cell Lymphoma: Cohort a of the Multicenter, Open-Label, Phase 2 Waveline-006 Study. Blood 2024, 144 (Suppl. 1), 4405. [Google Scholar] [CrossRef]
  45. Budde, L.E.; Matasar, M.; Sehn, L.H.; Assouline, S.E.; Kuruvilla, J.; Flinn, I.W.; Yoon, S.-S.; Bosch, F.; Greenwald, D.; Gutierrez, N.C.; et al. Mosunetuzumab Monotherapy Demonstrates Encouraging Activity and a Manageable Safety Profile in Patients with Heavily Pre-Treated Relapsed or Refractory Mantle Cell Lymphoma. Blood 2024, 144 (Suppl. 1), 1646. [Google Scholar] [CrossRef]
  46. Budde, L.E.; Olszewski, A.J.; Assouline, S.; Lossos, I.S.; Diefenbach, C.; Kamdar, M.; Ghosh, N.; Modi, D.; Sabry, W.; Naik, S.; et al. Mosunetuzumab with polatuzumab vedotin in relapsed or refractory aggressive large B cell lymphoma: A phase 1b/2 trial. Nat. Med. 2024, 30, 229–239. [Google Scholar] [CrossRef]
  47. Bannerji, R.; Arnason, J.E.; Advani, R.H.; Brown, J.R.; Allan, J.N.; Ansell, S.M.; Barnes, J.A.; O’Brien, S.M.; Chávez, J.C.; Duell, J.; et al. Odronextamab, a human CD20×CD3 bispecific antibody in patients with CD20-positive B-cell malignancies (ELM-1): Results from the relapsed or refractory non-Hodgkin lymphoma cohort in a single-arm, multicentre, phase 1 trial. Lancet Haematol. 2022, 9, e327–e339. [Google Scholar] [CrossRef]
  48. Hamadani, M.; Radford, J.; Carlo-Stella, C.; Caimi, P.F.; Reid, E.; O’Connor, O.A.; Feingold, J.M.; Ardeshna, K.M.; Townsend, W.; Solh, M.; et al. Final results of a phase 1 study of loncastuximab tesirine in relapsed/refractory B-cell non-Hodgkin lymphoma. Blood 2021, 137, 2634–2645. [Google Scholar] [CrossRef]
  49. Wang, M.; Munoz, J.; Goy, A.; Locke, F.L.; Jacobson, C.A.; Hill, B.T.; Timmerman, J.M.; Holmes, H.; Jaglowski, S.; Flinn, I.W.; et al. KTE-X19 CAR T-Cell Therapy in Relapsed or Refractory Mantle-Cell Lymphoma. N. Engl. J. Med. 2020, 382, 1331–1342. [Google Scholar] [CrossRef]
  50. Wang, M.; Munoz, J.; Goy, A.; Locke, F.L.; Jacobson, C.A.; Hill, B.T.; Timmerman, J.M.; Holmes, H.; Jaglowski, S.; Flinn, I.W.; 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, 555–567. [Google Scholar] [CrossRef]
  51. Wang, Y.; Jain, P.; Locke, F.L.; Maurer, M.J.; Frank, M.J.; Munoz, J.L.; Dahiya, S.; Beitinjaneh, A.M.; Jacobs, M.T.; McGuirk, J.P.; et al. Brexucabtagene Autoleucel for Relapsed or Refractory Mantle Cell Lymphoma in Standard-of-Care Practice: Results From the US Lymphoma CAR T Consortium. J. Clin. Oncol. 2023, 41, 2594–2606. [Google Scholar] [CrossRef] [PubMed]
  52. Wang, M.; Siddiqi, T.; Gordon, L.I.; Kamdar, M.; Lunning, M.; Hirayama, A.V.; Abramson, J.S.; Arnason, J.; Ghosh, N.; Mehta, A.; et al. Lisocabtagene Maraleucel in Relapsed/Refractory Mantle Cell Lymphoma: Primary Analysis of the Mantle Cell Lymphoma Cohort From TRANSCEND NHL 001, a Phase I Multicenter Seamless Design Study. J. Clin. Oncol. 2024, 42, 1146–1157. [Google Scholar] [CrossRef] [PubMed]
  53. Iacoboni, G.; Rejeski, K.; Villacampa, G.; van Doesum, J.A.; Chiappella, A.; Bonifazi, F.; Lopez-Corral, L.; van Aalderen, M.; Kwon, M.; Martínez-Cibrian, N.; et al. Real-world evidence of brexucabtagene autoleucel for the treatment of relapsed or refractory mantle cell lymphoma. Blood Adv. 2022, 6, 3606–3610. [Google Scholar] [CrossRef] [PubMed]
  54. Hess, G.; Dreyling, M.; Oberic, L.; Gine, E.; Zinzani, P.L.; Linton, K.; Vilmar, A.; Jerkeman, M.; Chen, J.M.H.; Ohler, A.; et al. Indirect treatment comparison of brexucabtagene autoleucel (ZUMA-2) versus standard of care (SCHOLAR-2) in relapsed/refractory mantle cell lymphoma. Leuk. Lymphoma 2024, 65, 14–25. [Google Scholar] [CrossRef]
  55. Ruan, J.; Martin, P.; Christos, P.; Cerchietti, L.; Tam, W.; Shah, B.; Schuster, S.J.; Rodriguez, A.; Hyman, D.; Calvo-Vidal, M.N.; et al. Five-year follow-up of lenalidomide plus rituximab as initial treatment of mantle cell lymphoma. Blood 2018, 132, 2016–2025. [Google Scholar] [CrossRef]
  56. Arcaini, L.; Lamy, T.; Walewski, J.; Belada, D.; Mayer, J.; Radford, J.; Jurczak, W.; Morschhauser, F.; Alexeeva, J.; Rule, S.; et al. Prospective subgroup analyses of the randomized MCL-002 (SPRINT) study: Lenalidomide versus investigator’s choice in relapsed or refractory mantle cell lymphoma. Br. J. Haematol. 2018, 180, 224–235. [Google Scholar] [CrossRef]
  57. Eve, H.E.; Carey, S.; Richardson, S.J.; Heise, C.C.; Mamidipudi, V.; Shi, T.; Radford, J.A.; Auer, R.L.; Bullard, S.H.; Rule, S.A. Single-agent lenalidomide in relapsed/refractory mantle cell lymphoma: Results from a UK phase II study suggest activity and possible gender differences. Br. J. Haematol. 2012, 159, 154–163. [Google Scholar] [CrossRef]
  58. Ruan, J.; Martin, P.; Shah, B.; Schuster, S.J.; Smith, S.M.; Furman, R.R.; Christos, P.; Rodriguez, A.; Svoboda, J.; Lewis, J.; et al. Lenalidomide plus Rituximab as Initial Treatment for Mantle-Cell Lymphoma. N. Engl. J. Med. 2015, 373, 1835–1844. [Google Scholar] [CrossRef]
  59. Nakhoda, S.; Vistarop, A.; Wang, Y.L. Resistance to Bruton tyrosine kinase inhibition in chronic lymphocytic leukaemia and non-Hodgkin lymphoma. Br. J. Haematol. 2023, 200, 137–149. [Google Scholar] [CrossRef]
  60. Das, D.; Wang, J.; Hong, J. Next-generation Bruton’s Tyrosine Kinase (BTK) Inhibitors Potentially Targeting BTK C481S Mutation- Recent Developments and Perspectives. Curr. Top. Med. Chem. 2022, 22, 1674–1691. [Google Scholar] [CrossRef]
  61. Mato, A.R.; Flinn, I.W.; Pagel, J.M.; Brown, J.R.; Cheah, C.Y.; Coombs, C.C.; Patel, M.R.; Rothenberg, S.M.; Tsai, D.E.; Ku, N.C.; et al. Results from a First-in-Human, Proof-of-Concept Phase 1 Trial in Pretreated B-Cell Malignancies for Loxo-305, a Next-Generation, Highly Selective, Non-Covalent BTK Inhibitor. Blood 2019, 134 (Suppl. 1), 501. [Google Scholar] [CrossRef]
  62. Telaraja, D.; Kasamon, Y.L.; Collazo, J.S.; Leong, R.; Wang, K.; Li, P.; Dahmane, E.; Yang, Y.; Earp, J.; Grimstein, M.; et al. FDA Approval Summary: Pirtobrutinib for Relapsed or Refractory Mantle Cell Lymphoma. Clin. Cancer Res. 2024, 30, 17–22. [Google Scholar] [CrossRef] [PubMed]
  63. Eyre, T.A.; Shah, N.N.; Dreyling, M.; Jurczak, W.; Wang, Y.; Cheah, C.Y.; Song, Y.; Gandhi, M.; Chay, C.; Sharman, J.; et al. BRUIN MCL-321: Phase III study of pirtobrutinib versus investigator choice of BTK inhibitor in BTK inhibitor naive mantle cell lymphoma. Future Oncol. 2022, 18, 3961–3969. [Google Scholar] [CrossRef]
  64. VENCLEXTA (Venetoclax) [Prescribing Information]; AbbVie Inc.: North Chicago, IL, USA,, 2019.
  65. Haselager, M.V.; Kielbassa, K.; Ter Burg, J.; Bax, D.J.C.; Fernandes, S.M.; Borst, J.; Tam, C.; Forconi, F.; Chiodin, G.; Brown, J.R.; et al. Changes in Bcl-2 members after ibrutinib or venetoclax uncover functional hierarchy in determining resistance to venetoclax in CLL. Blood 2020, 136, 2918–2926. [Google Scholar] [CrossRef]
  66. Wang, M.; Ramchandren, R.; Chen, R.; Karlin, L.; Chong, G.; Jurczak, W.; Wu, K.L.; Bishton, M.; Collins, G.P.; Eliadis, P.; et al. Concurrent ibrutinib plus venetoclax in relapsed/refractory mantle cell lymphoma: The safety run-in of the phase 3 SYMPATICO study. J. Hematol. Oncol. 2021, 14, 179. [Google Scholar] [CrossRef]
  67. Wang, M.; Jurczak, W.; Trněný, M.; Belada, D.; Wrobel, T.; Ghosh, N.; Keating, M.-M.; van Meerten, T.; Fernandez Alvarez, R.; von Keudell, G.; et al. Ibrutinib Combined with Venetoclax in Patients with Relapsed/Refractory Mantle Cell Lymphoma: Primary Analysis Results from the Randomized Phase 3 Sympatico Study. Blood 2023, 142 (Suppl. 2), LBA-2. [Google Scholar] [CrossRef]
  68. Wang, M.; Jurczak, W.; Trneny, M.; Belada, D.; Wrobel, T.; Ghosh, N.; Keating, M.M.; van Meerten, T.; Alvarez, R.F.; von Keudell, G.; et al. Ibrutinib plus venetoclax in relapsed or refractory mantle cell lymphoma (SYMPATICO): A multicentre, randomised, double-blind, placebo-controlled, phase 3 study. Lancet Oncol. 2025, 26, 200–213. [Google Scholar] [CrossRef]
  69. Spurgeon, S.; Mei, M.; Barr, P.; Barrientos, J.; Vos, S.; Furman, R.; Patel, K.; Thompson, P.; Choi, M.; Kallam, A.; et al. Waveline-001: Updated Results from a Phase 1 Dose Escalation and Cohort Expansion Study of Zilovertamab Vedotin (MK-2140) in Non-Hodgkin Lymphoma. Blood 2022, 140, 6640–6641. [Google Scholar] [CrossRef]
  70. Wang, M.L.; Barrientos, J.C.; Furman, R.R.; Mei, M.; Barr, P.M.; Choi, M.Y.; de Vos, S.; Kallam, A.; Patel, K.; Kipps, T.J.; et al. Zilovertamab Vedotin Targeting of ROR1 as Therapy for Lymphoid Cancers. NEJM Evid. 2022, 1, EVIDoa2100001. [Google Scholar] [CrossRef]
  71. Calabretta, E.; Hamadani, M.; Zinzani, P.L.; Caimi, P.; Carlo-Stella, C. The antibody-drug conjugate loncastuximab tesirine for the treatment of diffuse large B-cell lymphoma. Blood 2022, 140, 303–308. [Google Scholar] [CrossRef]
  72. Liu, D. Cancer biomarkers for targeted therapy. Biomark. Res. 2019, 7, 25. [Google Scholar] [CrossRef] [PubMed]
  73. Caimi, P.F.; Ai, W.; Alderuccio, J.P.; Ardeshna, K.M.; Hamadani, M.; Hess, B.; Kahl, B.S.; Radford, J.; Solh, M.; Stathis, A.; et al. Loncastuximab tesirine in relapsed or refractory diffuse large B-cell lymphoma (LOTIS-2): A multicentre, open-label, single-arm, phase 2 trial. Lancet Oncol. 2021, 22, 790–800. [Google Scholar] [CrossRef] [PubMed]
  74. Consolidation With Loncastuximab Tesirine After a Short Course of Immunochemotherapy in BTKi-Treated (or Intolerant) Relapsed/Refractory Mantle Cell Lymphoma Patients. (FIL_COLUMN) ClinicalTrials.gov ID NCT05249959. Available online: https://inclinicaltrials.com/refractory-mantle-cell-lymphoma/NCT05249959/ (accessed on 26 June 2025).
  75. Depaus, J.; Bryan, L.; Ungar, D.; Dautaj, I.; Wagner-Johnston, N. Safety and Anti-Tumor Activity Study of Loncastuximab Tesirine and Ibrutinib in Diffuse Large B-Cell or Mantle Cell Lymphoma. Blood 2019, 134, 5309. [Google Scholar] [CrossRef]
  76. Moskowitz, C.H.; Bastos-Oreiro, M.; Ungar, D.; Dautaj, I.; Kalac, M. Safety and Anti-Tumor Activity Study of Loncastuximab Tesirine and Durvalumab in Diffuse Large B-Cell, Mantle Cell, or Follicular Lymphoma. Blood 2019, 134 (Suppl. 1), 2807. [Google Scholar] [CrossRef]
  77. Bacac, M.; Colombetti, S.; Herter, S.; Sam, J.; Perro, M.; Chen, S.; Bianchi, R.; Richard, M.; Schoenle, A.; Nicolini, V.; et al. CD20-TCB with Obinutuzumab Pretreatment as Next-Generation Treatment of Hematologic Malignancies. Clin. Cancer Res. 2018, 24, 4785–4797. [Google Scholar] [CrossRef]
  78. COLUMVI Prescribing Information. Available online: https://www.gene.com/download/pdf/columvi_prescribing.pdf (accessed on 30 June 2025).
  79. Phillips, T.J.; Carlo-Stella, C.; Morschhauser, F.; Bachy, E.; Crump, M.; Trněný, M.; Bartlett, N.L.; Zaucha, J.; Wrobel, T.; Offner, F.; et al. Glofitamab in Relapsed/Refractory Mantle Cell Lymphoma: Results From a Phase I/II Study. J. Clin. Oncol. 2025, 43, 318–328. [Google Scholar] [CrossRef]
  80. Bröske, A.E.; Korfi, K.; Belousov, A.; Wilson, S.; Ooi, C.H.; Bolen, C.R.; Canamero, M.; Alcaide, E.G.; James, I.; Piccione, E.C.; et al. Pharmacodynamics and molecular correlates of response to glofitamab in relapsed/refractory non-Hodgkin lymphoma. Blood Adv. 2022, 6, 1025–1037. [Google Scholar] [CrossRef]
  81. Phillips, T.J.; Matasar, M.; Eyre, T.A.; Gine, E.; Filézac De L’Étang, A.; Byrne, B.; Lundberg, L.; Padovani, A.; Boetsch, C.; Bottos, A.; et al. GLOBRYTE: A Phase III, Open-Label, Multicenter, Randomized Trial Evaluating Glofitamab Monotherapy in Patients with Relapsed or Refractory Mantle Cell Lymphoma. Blood 2023, 142 (Suppl. 1), 3052. [Google Scholar] [CrossRef]
  82. Engelberts, P.J.; Hiemstra, I.H.; de Jong, B.; Schuurhuis, D.H.; Meesters, J.; Beltran Hernandez, I.; Oostindie, S.C.; Neijssen, J.; van den Brink, E.N.; Horbach, G.J.; et al. DuoBody-CD3xCD20 induces potent T-cell-mediated killing of malignant B cells in preclinical models and provides opportunities for subcutaneous dosing. EBioMedicine 2020, 52, 102625. [Google Scholar] [CrossRef]
  83. Labrijn, A.F.; Meesters, J.I.; de Goeij, B.E.; van den Bremer, E.T.; Neijssen, J.; van Kampen, M.D.; Strumane, K.; Verploegen, S.; Kundu, A.; Gramer, M.J.; et al. Efficient generation of stable bispecific IgG1 by controlled Fab-arm exchange. Proc. Natl. Acad. Sci. USA 2013, 110, 5145–5150. [Google Scholar] [CrossRef]
  84. Labrijn, A.F.; Meesters, J.I.; Priem, P.; de Jong, R.N.; van den Bremer, E.T.; van Kampen, M.D.; Gerritsen, A.F.; Schuurman, J.; Parren, P.W. Controlled Fab-arm exchange for the generation of stable bispecific IgG1. Nat. Protoc. 2014, 9, 2450–2463. [Google Scholar] [CrossRef] [PubMed]
  85. Hutchings, M.; Mous, R.; Clausen, M.R.; Johnson, P.; Linton, K.M.; Chamuleau, M.E.D.; Lewis, D.J.; Sureda Balari, A.; Cunningham, D.; Oliveri, R.S.; et al. Dose escalation of subcutaneous epcoritamab in patients with relapsed or refractory B-cell non-Hodgkin lymphoma: An open-label, phase 1/2 study. Lancet 2021, 398, 1157–1169. [Google Scholar] [CrossRef] [PubMed]
  86. Budde, L.E.; Assouline, S.; Sehn, L.H.; Schuster, S.J.; Yoon, S.S.; Yoon, D.H.; Matasar, M.J.; Bosch, F.; Kim, W.S.; Nastoupil, L.J.; et al. Durable Responses With Mosunetuzumab in Relapsed/Refractory Indolent and Aggressive B-Cell Non-Hodgkin Lymphomas: Extended Follow-Up of a Phase I/II Study. J. Clin. Oncol. 2024, 42, 2250–2256. [Google Scholar] [CrossRef] [PubMed]
  87. Schuster, S.J.; Sehn, L.H.; Bartlett, N.L.; Matasar, M.; Assouline, S.; Giri, P.; Kuruvilla, J.; Shadman, M.; Cheah, C.Y.; Dietrich, S.; et al. Mosunetuzumab Monotherapy Continues to Demonstrate Durable Responses in Patients with Relapsed and/or Refractory Follicular Lymphoma after ≥2 Prior Therapies: 3-Year Follow-up from a Pivotal Phase II Study. Blood 2023, 142 (Suppl. 1), 603. [Google Scholar] [CrossRef]
  88. Budde, L.E.; Sehn, L.H.; Matasar, M.; Schuster, S.J.; Assouline, S.; Giri, P.; Kuruvilla, J.; Canales, M.; Dietrich, S.; Fay, K.; et al. Safety and efficacy of mosunetuzumab, a bispecific antibody, in patients with relapsed or refractory follicular lymphoma: A single-arm, multicentre, phase 2 study. Lancet Oncol. 2022, 23, 1055–1065. [Google Scholar] [CrossRef]
  89. Budde, E.L.; Bartlett, N.L.; Giri, P.; Schuster, S.J.; Assouline, S.; Yoon, S.-S.; Fay, K.; Matasar, M.J.; Gutierrez, N.C.; Marlton, P.; et al. Subcutaneous Mosunetuzumab Is Active with a Manageable Safety Profile in Patients (pts) with Relapsed/Refractory (R/R) B-Cell Non-Hodgkin Lymphomas (B-NHLs): Updated Results from a Phase I/II Study. Blood 2022, 140 (Suppl. 1), 3753–3755. [Google Scholar] [CrossRef]
  90. Available online: https://www.ema.europa.eu/en/medicines/human/EPAR/lunsumio#authorisation-details (accessed on 26 June 2025).
  91. Budde, L.E.; Assouline, S.; Sehn, L.H.; Schuster, S.J.; Yoon, S.S.; Yoon, D.H.; Matasar, M.J.; Bosch, F.; Kim, W.S.; Nastoupil, L.J.; et al. Single-Agent Mosunetuzumab Shows Durable Complete Responses in Patients With Relapsed or Refractory B-Cell Lymphomas: Phase I Dose-Escalation Study. J. Clin. Oncol. 2022, 40, 481–491. [Google Scholar] [CrossRef]
  92. Bartlett, N.L.; Assouline, S.; Giri, P.; Schuster, S.J.; Cheah, C.Y.; Matasar, M.; Gregory, G.P.; Yoon, D.H.; Shadman, M.; Fay, K.; et al. Mosunetuzumab monotherapy is active and tolerable in patients with relapsed/refractory diffuse large B-cell lymphoma. Blood Adv. 2023, 7, 4926–4935. [Google Scholar] [CrossRef]
  93. Lynch, R.C.; Poh, C.; Shadman, M.; Till, B.G.; Smith, S.D.; Ujjani, C.S.; Ottemiller, S.; Fessel, M.M.; Joy, B.; DeBell, J.; et al. Early Complete Responses with Mosunetuzumab Monotherapy in Treatment-Naïve Follicular and Marginal Zone Lymphomas with Only Low-Grade Cytokine Release Syndrome. Blood 2023, 142 (Suppl. 1), 4397. [Google Scholar] [CrossRef]
  94. Cheah, C.Y.; Assouline, S.; Baker, R.; Bartlett, N.L.; El-Sharkawi, D.; Giri, P.; Ku, M.; Schuster, S.J.; Matasar, M.; Radford, J.; et al. Mosunetuzumab Monotherapy Demonstrates Activity and a Manageable Safety Profile in Patients with Relapsed or Refractory Richter’s Transformation. Blood 2023, 142 (Suppl. 1), 614. [Google Scholar] [CrossRef]
  95. Wang, M.L.; Assouline, S.; Kamdar, M.; Ghosh, N.; Naik, S.; Nakhoda, S.K.; Chavez, J.C.; Jia, T.; Pham, S.; Huw, L.-Y.; et al. Fixed Duration Mosunetuzumab Plus Polatuzumab Vedotin Has Promising Efficacy and a Manageable Safety Profile in Patients with BTKi Relapsed/Refractory Mantle Cell Lymphoma: Initial Results from a Phase Ib/II Study. Blood 2023, 142 (Suppl. 1), 734. [Google Scholar] [CrossRef]
  96. Kim, W.S.; Kim, T.M.; Cho, S.G.; Jarque, I.; Iskierka-Jażdżewska, E.; Poon, L.M.; Prince, H.M.; Zhang, H.; Cao, J.; Zhang, M.; et al. Odronextamab monotherapy in patients with relapsed/refractory diffuse large B cell lymphoma: Primary efficacy and safety analysis in phase 2 ELM-2 trial. Nat. Cancer 2025, 6, 528–539. [Google Scholar] [CrossRef] [PubMed]
  97. Ayyappan, S.; Kim, W.S.; Kim, T.M.; Walewski, J.; Cho, S.-G.; Jarque, I.; Iskierka-Jazdzewska, E.; Poon, M.; Oh, S.Y.; Inng Lim, F.L.W.; et al. Final Analysis of the Phase 2 ELM-2 Study: Odronextamab in Patients with Relapsed/Refractory (R/R) Diffuse Large B-Cell Lymphoma (DLBCL). Blood 2023, 142, 436. [Google Scholar] [CrossRef]
  98. Brouwer-Visser, J.; Fiaschi, N.; Deering, R.P.; Cygan, K.J.; Scott, D.; Jeong, S.; Boucher, L.; Gupta, N.T.; Gupta, S.; Adler, C.; et al. Molecular assessment of intratumoral immune cell subsets and potential mechanisms of resistance to odronextamab, a CD20×CD3 bispecific antibody, in patients with relapsed/refractory B-cell non-Hodgkin lymphoma. J. Immunother. Cancer 2024, 12, e008338. [Google Scholar] [CrossRef]
  99. Martino, M.; Canale, F.A.; Naso, V.; Porto, G.; Gerace, D.; Allegra, A. Do CAR-T and Allogeneic Stem Cell Transplant Both Have a Place in Lymphoid Neoplasms? Int. J. Mol. Sci. 2023, 24, 1045. [Google Scholar] [CrossRef] [PubMed]
  100. Gutierrez, A.; Bento, L.; Novelli, S.; Martin, A.; Gutierrez, G.; Queralt Salas, M.; Bastos-Oreiro, M.; Perez, A.; Hernani, R.; Cruz Viguria, M.; et al. Allogeneic Stem Cell Transplantation in Mantle Cell Lymphoma; Insights into Its Potential Role in the Era of New Immunotherapeutic and Targeted Therapies: The GETH/GELTAMO Experience. Cancers 2022, 14, 2673. [Google Scholar] [CrossRef]
  101. Di Blasi, R.; Le Gouill, S.; Bachy, E.; Cartron, G.; Beauvais, D.; Le Bras, F.; Gros, F.X.; Choquet, S.; Bories, P.; Feugier, P.; et al. Outcomes of patients with aggressive B-cell lymphoma after failure of anti-CD19 CAR T-cell therapy: A DESCAR-T analysis. Blood 2022, 140, 2584–2593. [Google Scholar] [CrossRef]
  102. Liebers, N.; Boumendil, A.; Finel, H.; Edelmann, D.; Kobbe, G.; Baermann, B.N.; Serroukh, Y.; Blaise, D.; Beelen, D.W.; Solano, C.; et al. Brexucabtagene autoleucel versus allogeneic hematopoietic cell transplantation in relapsed and refractory mantle cell lymphoma. Blood Cancer Discov. 2025, 6, 182–190. [Google Scholar] [CrossRef]
  103. Zurko, J.; Nizamuddin, I.; Epperla, N.; David, K.; Cohen, J.B.; Moyo, T.K.; Ollila, T.; Hess, B.; Roy, I.; Ferdman, R.; et al. Peri-CAR-T practice patterns and survival predictors for all CAR-T patients and post-CAR-T failure in aggressive B-NHL. Blood Adv. 2023, 7, 2657–2669. [Google Scholar] [CrossRef]
  104. Fried, S.; Shouval, R.; Walji, M.; Flynn, J.R.; Yerushalmi, R.; Shem-Tov, N.; Danylesko, I.; Tomas, A.A.; Fein, J.A.; Devlin, S.M.; et al. Allogeneic Hematopoietic Cell Transplantation after Chimeric Antigen Receptor T Cell Therapy in Large B Cell Lymphoma. Transplant. Cell Ther. 2023, 29, 99–107. [Google Scholar] [CrossRef]
  105. Iacoboni, G.; Iraola-Truchuelo, J.; O’Reilly, M.; Navarro, V.; Menne, T.; Kwon, M.; Martín-López, A.; Chaganti, S.; Delgado, J.; Roddie, C.; et al. Treatment outcomes in patients with large B-cell lymphoma after progression to chimeric antigen receptor T-cell therapy. Hemasphere 2024, 8, e62. [Google Scholar] [CrossRef] [PubMed]
  106. Montoya, S.; Bourcier, J.; Noviski, M.; Lu, H.; Thompson, M.C.; Chirino, A.; Jahn, J.; Sondhi, A.K.; Gajewski, S.; Tan, Y.S.M.; et al. Kinase-impaired BTK mutations are susceptible to clinical-stage BTK and IKZF1/3 degrader NX-2127. Science 2024, 383, eadi5798. [Google Scholar] [CrossRef] [PubMed]
  107. Danilov, A.; Tees, M.; Patel, K.; Wierda, W.; Patel, M.; Flinn, I.; Latif, T.; Ai, W.; Thompson, M.; Wang, M.; et al. A First-in-Human Phase 1 Trial of NX-2127, a First-in-Class Bruton’s Tyrosine Kinase (BTK) Dual-Targeted Protein Degrader with Immunomodulatory Activity, in Patients with Relapsed/Refractory B Cell Malignancies. Blood 2023, 142, 4463. [Google Scholar] [CrossRef]
  108. Shah, N.N.; Omer, Z.; Collins, G.P.; Forconi, F.; Danilov, A.; Byrd, J.C.; El-Sharkawi, D.; Searle, E.; Alencar, A.J.; Ma, S.; et al. Efficacy and Safety of the Bruton’s Tyrosine Kinase (BTK) Degrader NX-5948 in Patients with Relapsed/Refractory (R/R) Chronic Lymphocytic Leukemia (CLL): Updated Results from an Ongoing Phase 1a/b Study. Blood 2024, 144 (Suppl. 1), 884. [Google Scholar] [CrossRef]
  109. Profitos-Peleja, N. The Multikinase CDK4/6 Inhibitor Narazaciclib Overcomes Btki Resistance in Mantle Cell Lymphoma by Targeting USP24-P53. Available online: https://www.researchgate.net/publication/371455714_Prolonged_cell_cycle_arrest_by_the_CDK46_antagonist_narazaciclib_restores_ibrutinib_response_in_preclinical_models_of_BTKi-resistant_mantle_cell_lymphoma (accessed on 26 June 2025).
  110. Parra, J.R.M.; et al. (Eds.) Hemasphere, 2024; Volume 8. (Suppl. 1), 1272.
  111. Lantermans, H.C.; Ma, F.; Kuil, A.; van Kesteren, S.; Yasinoglu, S.; Yang, G.; Buhrlage, S.J.; Wang, J.; Gray, N.S.; Kersten, M.J.; et al. The dual HCK/BTK inhibitor KIN-8194 impairs growth and integrin-mediated adhesion of BTKi-resistant mantle cell lymphoma. Leukemia 2024, 38, 1570–1580. [Google Scholar] [CrossRef]
  112. Durand, R.; Bellanger, C.; Kervoëlen, C.; Tessoulin, B.; Dousset, C.; Menoret, E.; Asnagli, H.; Parker, A.; Beer, P.; Pellat-Deceunynck, C.; et al. Selective pharmacologic targeting of CTPS1 shows single-agent activity and synergizes with BCL2 inhibition in aggressive mantle cell lymphoma. Haematologica 2024, 109, 2574–2584. [Google Scholar] [CrossRef]
  113. Thus, Y.J.; Eldering, E.; Kater, A.P.; Spaargaren, M. Tipping the balance: Toward rational combination therapies to overcome venetoclax resistance in mantle cell lymphoma. Leukemia 2022, 36, 2165–2176. [Google Scholar] [CrossRef]
  114. Dolnikova, A.; Kazantsev, D.; Klanova, M.; Pokorna, E.; Sovilj, D.; Kelemen, C.D.; Tuskova, L.; Hoferkova, E.; Mraz, M.; Helman, K.; et al. Blockage of BCL-XL overcomes venetoclax resistance across BCL2+ lymphoid malignancies irrespective of BIM status. Blood Adv. 2024, 8, 3532–3543. [Google Scholar] [CrossRef]
  115. Goy, A.; Hernandez-Ilzaliturri, F.J.; Kahl, B.; Ford, P.; Protomastro, E.; Berger, M. A phase I/II study of the pan Bcl-2 inhibitor obatoclax mesylate plus bortezomib for relapsed or refractory mantle cell lymphoma. Leuk. Lymphoma 2014, 55, 2761–2768. [Google Scholar] [CrossRef]
  116. Cheah, C.; Tam, C.; Lasica, M.; Verner, E.; Browett, P.; Anderson, M.A.; Hilger, J.; Fang, Y.; Simpson, D.; Opat, S. A Phase 1 Study with the Novel B-Cell Lymphoma 2 (Bcl-2) Inhibitor Bgb-11417 As Monotherapy or in Combination with Zanubrutinib (ZANU) in Patients (Pts) with CLL/SLL: Preliminary Data. Blood 2022, 140, 2321–2323. [Google Scholar] [CrossRef]
  117. Soumerai, J.; Lasica, M.; Opat, S.; Cheah, C.; Chan, H.; Verner, E.; Barca, E.; Tedeschi, A.; Hilger, J.; Fang, Y.; et al. A Phase 1 Study with the Novel B-Cell Lymphoma 2 (Bcl-2) Inhibitor Bgb-11417 As Monotherapy or in Combination with Zanubrutinib (ZANU) in Patients (Pts) with Non-Hodgkin Lymphoma (NHL) or Waldenström Macroglobulinemia (WM): Preliminary Data. Blood 2022, 140, 9325–9327. [Google Scholar] [CrossRef]
  118. Hodson, D.; Shouse, G.; Shin, H.J.; Salar Silvestre, A.; Bobillo Varela, S.; Ribrag, V.; Macpherson, N.; Cordoba, R.; Seok Kim, J.; Radford, J.; et al. P1098: A Phase II, Open-Label, Multicenter Study of Capivasertib, A Potent, Oral Pan-AKT Inhibitor, in Patients with Relapsed or Refractory B-Cell Non-Hodgkin Lymphoma (Capital). Hemasphere 2023, 7, e5749555. [Google Scholar] [CrossRef]
  119. Wang, M.; Johnson, P.; Yazji, S.; Katz, Y.; Pietrofeso, A.; Robinson, J.; Mei, M.; Frigault, M.; Breitmeyer, J.; Jacobson, C. A Phase 1/2 Study of a ROR1-Targeting CAR T Cell Therapy (ONCT-808) in Adult Patients with Relapsed/Refractory (R/R) Aggressive B Cell Lymphomas (BCL). Blood 2024, 144, 1743. [Google Scholar] [CrossRef]
  120. Shah, N.N.; Fenske, T.; Johnson, B.; Szabo, A.; Devata, S.; Longo, W.; Kearl, T.; Zamora, A.; Hari, P.; Schneider, D.; et al. P1082: Results from a Phase 1/2 Study of Tandem, Bispecific Anti-CD20/Anti-CD19 (LV20.19) CAR T-Cells for Mantle Cell Lymphoma. Hemasphere 2023, 7, e207658e. [Google Scholar] [CrossRef]
  121. Chan, W.K.; Williams, J.; Sorathia, K.; Pray, B.; Abusaleh, K.; Bian, Z.; Sharma, A.; Hout, I.; Nishat, S.; Hanel, W.; et al. A novel CAR-T cell product targeting CD74 is an effective therapeutic approach in preclinical mantle cell lymphoma models. Exp. Hematol. Oncol. 2023, 12, 79. [Google Scholar] [CrossRef]
Figure 1. Main therapeutic targets and active drugs under investigation in mantle cell lymphoma.
Figure 1. Main therapeutic targets and active drugs under investigation in mantle cell lymphoma.
Cancers 17 02239 g001
Figure 2. Proposed therapeutic algorithm for mantle cell lymphoma including both clinically available and experimental drugs, expected in the near future. Abbreviations—BR: bendamustine rituximab; BCL: B-cell lymphoma; BTKi: Bruton tyrosine kinase inhibitor; CAR-T: chimeric antigen receptor-T cell; R: rituximab.
Figure 2. Proposed therapeutic algorithm for mantle cell lymphoma including both clinically available and experimental drugs, expected in the near future. Abbreviations—BR: bendamustine rituximab; BCL: B-cell lymphoma; BTKi: Bruton tyrosine kinase inhibitor; CAR-T: chimeric antigen receptor-T cell; R: rituximab.
Cancers 17 02239 g002
Table 1. Comparison of covalent Bruton Tyrosine Kinase inhibitors in mantle cell lymphoma.
Table 1. Comparison of covalent Bruton Tyrosine Kinase inhibitors in mantle cell lymphoma.
DrugStudyPatient NumberPost-ASCTPost-CAR-TORRCROSmPFSmDORmFUAEs ≥ G3Ref
IbrutinibNCT0123639111112 (11%)-68%21%N.R.13.917.515.3 [22]
IbrutinibNCT01646021280--72%19%N.R.14.6N.R.2094 (68%)[23]
AcalabrutinibNCT0221392612422 (18%)-81%40%N.R.N.R.N.R.15.248 (39%)[24]
ZanubrutinibNCT0320697086--83.7%77.9%N.R.33N.R.35.3 [30]
Abbreviations—AEs ≥ G3: adverse events of grade equal or above 3; ASCT: autologous stem cell transplant; CAR-T: chimeric antigen receptor-T cell; CR: complete response; mDOR: median duration of response; mFU: median follow-up; mPFS: median progression-free survival; NR: not reached; ORR: overall response rate; OS: overall survival.
Table 2. Available data on potential drugs in patients with R/R MCL after CAR-T failure or in patients ineligible for CAR-T.
Table 2. Available data on potential drugs in patients with R/R MCL after CAR-T failure or in patients ineligible for CAR-T.
StudyPatient NumberPost-ASCTPost-CAR-TORRCROSmPFSmDORmFUAEs ≥ G3Ref
PirtobrutinibNCT03740529127 81%43%NR 62% at 18 monthsNR 74% at 18 months 18(20.5%)[41]
Venetoclax + ibrutinibNCT0311217426739 (29%) 75%21%36.731.9 (84%)[42]
GlofitamabNCT03075696612 (3.3%)16 (26.7%)74%71%29.916.816.219.6(65%)[43]
Zilovertamab vedotinNCT038331804011 (28%)6 (15%)40%13%93.43 32 (80%)[44]
MosunetuzumabNCT0250040725(33%) 44%24%7.33.710.354.519 (76%)[45]
Mosunetuzumab + polatuzumabNCT036710182015 (12.5%)42 (35%)75%70%23.311.4NR at mFU of 45 months45(56.7%)[46]
OdronextamabNCT0229095112 3 (25%)50%33% 7.626.2 [47]
Loncastuximab-tesirineNCT0266901715 46.7%33%N.R.4.8 [48]
Abbreviations—AEs ≥ G3: adverse events of grade equal or above 3; ASCT: autologous stem cell transplant; CAR-T: chimeric antigen receptor-T cell; CR: complete response; mDOR: median duration of response; mFU: median follow-up; mPFS: median progression-free survival; NR: not reached; ORR: overall response rate; OS: overall survival.
Table 3. Ongoing trials after BTKi and CAR-T in R/R MCL.
Table 3. Ongoing trials after BTKi and CAR-T in R/R MCL.
NCT NumberNamePhaseStateDrug
NCT04939272 I–IINot recruitingCopanlisib Hydrochloride, Venetoclax
NCT03891355FIL_KLIMTIINot recruitingCarfilzomib, Lenalidomide, Dexamethasone
NCT04047797 IINot recruitingIxazomib, Ixazomib Citrate, Rituximab
NCT02558816OAslsI–IINot recruitingIbrutinib, Obinutuzumab, GDC-0199 (Venetoclax)
NCT02446236 IbNot recruitingLenalidomide, Ibrutinib, Rituximab
NCT01695941 INot recruitingAlisertib, Bortezomib, Rituximab
NCT03440567 INot recruitingAutologous Hematopoietic Stem Cell Transplantation, Avelumab, Carboplatin, Etoposide Phosphate, Ibrutinib, Ifosfamide, Rituximab, Utomilumab
NCT04659044 IINot recruitingPolatuzumab Vedotin, Rituximab, Rituximab and Hyaluronidase Human, Venetoclax
NCT01996865MAGNIFYIIIbNot recruitingLenalidomide, Rituximab
NCT04703686 IINot recruitingObinutuzumab, RO7082859 (Glofitamab)
NCT03162536 I–IINot recruitingNemtabrutinib
NCT03015896 I–IINot recruitingLenalidomide, Nivolumab
NCT04578600 I/IbNot recruitingLenalidomide, Obinutuzumab, Oral Azacitidine
NCT03277729 I/IINot recruitingChimeric Antigen Receptor-T-Cell Therapy, Cyclophosphamide, Fludarabine Phosphate
NCT04205409 IINot recruitingNivolumab
NCT02568553 INot recruitingBlinatumomab, Lenalidomide
NCT03997968 I/IINot recruitingCYT-0851, CYT-0851 + gemcitabine/capecitabine/rituximab and bendamustine
NCT05131022 Ia/IbRecruitingNX5948
NCT06252675 IIRecruitingObinutuzumab, Glofitamab, Pirtobrutinib
NCT05471843 I/IINot recruitingBGB-11417
NCT05868395 IIRecruitingPolatuzumab, Bendamustine, Rituximab
NCT06324994 IINot yet recruitingLinperlisib + Obinutuzumab and Venetoclax
NCT06106841 Ib/IIRecruitingTQB3909
NCT04484012 IIRecruitingAcalabrutinib, CD19CAR-CD28-CD3zeta-EGFRt-expressing Tn/mem-enriched T lymphocytes
NCT04186520 I/IIRecruitingLV20.19 CAR-T
NCT05910801 IIRecruitingLenalidomide, Tafasitamab, Venetoclax
NCT05716087 IINot recruitingLP-168 (Rocbrutinib)
NCT06192888 IRecruitingGlofitamab, Obinutuzumab, Lenalidomide
NCT06558604GLOASISIIRecruitingObinutuzumab, Glofitamab, Venetoclax, Zanubrutinib
NCT05249959COLUMNIIRecruitingADCT-402 (loncastuximab tesirine), Rituximab-Bendamustine, Ara-C
NCT05529069 IIRecruitingPirtobrutinib, Venetoclax
NCT05444322 IRecruitingRD14-01 cell infusion (anti ROR1 CAR-T)
NCT06300528 IIRecruitingPemigatinib
NCT05990465 IRecruitingPirtobrutinib, LV20.19 CAR-T
NCT06208735 IRecruitingCLIC-2201
NCT04763083 IRecruitingNVG-111
NCT05370430 IRecruitingBAFFR-CAR-T cells
NCT06464861 IRecruitingCD19-CAR-NK/T
NCT06544265 IRecruitingSynKIR-310 (Autologous T Cells Transduced with CD19 KIR-CAR)
NCT03676504 I/IIRecruitingCD19.CAR-T Cells, Fludarabine, Cyclophosphamide
NCT04775745 IRecruitingLP-168 (Rocbrutinib)
NCT05643742 I/IIRecruitingCTX112 (Allogeneic CRISPR-Cas9 Engineered CD19 CAR-T)
NCT05453396 IIRecruitingLoncastuximab Tesirine
NCT06285422 IRecruitingSC262
NCT05887167 IRecruitingAutologous Stem Cell Transplantation, CAR-T
NCT04830137 Ia/IbRecruitingNX-2127
NCT02952508CLOVER-1IINot recruitingCLR 131 (Iopofosine I 131)
NCT06191887 Ia/IbRecruitingAutologous BAFFR-targeting CAR-T Cells, Bendamustine, Cyclophosphamide, Fludarabine
NCT06561425 I/IIRecruitingGLPG5101
NCT03888105 IIRecruitingOdronextamab
NCT03547115 IRecruitingVoruciclib
NCT06256484 IRecruitingATA3219
NCT04223765 IRecruitingCAR.k.28, Fludarabine, Cyclophosphamide, Bendamustine
NCT05780034 IRecruitingAC676
NCT06542250TITANiumI/IIRecruitingAZD5492
NCT05665530 INot recruitingPRT2527, Zanubrutinib, Venetoclax
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Boccellato, E.; Comba, L.; Tavarozzi, R.; Castellino, C.; Foglietta, M.; Mattei, D.; Ladetto, M.; Massaia, M.; Castellino, A. Next-Generation Therapies in Mantle Cell Lymphoma (MCL): The Evolving Landscape in Treatment of Relapse/Refractory After CAR-T Cells. Cancers 2025, 17, 2239. https://doi.org/10.3390/cancers17132239

AMA Style

Boccellato E, Comba L, Tavarozzi R, Castellino C, Foglietta M, Mattei D, Ladetto M, Massaia M, Castellino A. Next-Generation Therapies in Mantle Cell Lymphoma (MCL): The Evolving Landscape in Treatment of Relapse/Refractory After CAR-T Cells. Cancers. 2025; 17(13):2239. https://doi.org/10.3390/cancers17132239

Chicago/Turabian Style

Boccellato, Elia, Lorenzo Comba, Rita Tavarozzi, Claudia Castellino, Myriam Foglietta, Daniele Mattei, Marco Ladetto, Massimo Massaia, and Alessia Castellino. 2025. "Next-Generation Therapies in Mantle Cell Lymphoma (MCL): The Evolving Landscape in Treatment of Relapse/Refractory After CAR-T Cells" Cancers 17, no. 13: 2239. https://doi.org/10.3390/cancers17132239

APA Style

Boccellato, E., Comba, L., Tavarozzi, R., Castellino, C., Foglietta, M., Mattei, D., Ladetto, M., Massaia, M., & Castellino, A. (2025). Next-Generation Therapies in Mantle Cell Lymphoma (MCL): The Evolving Landscape in Treatment of Relapse/Refractory After CAR-T Cells. Cancers, 17(13), 2239. https://doi.org/10.3390/cancers17132239

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop