Abstract
Mantle cell lymphoma (MCL) is a heterogeneous aggressive disease and remains incurable with current chemotherapies. The development of monoclonal antibody (mAb) has led to substantial achievement in immunotherapeutic strategies for B-cell lymphomas including MCL. Nonetheless, progress in the clinical use of mAbs is hindered by poor efficacy, off-target toxicities and drug resistance. Thus, novel mAbs engineering and approaches to improve target specificity and enhance affinity and potency are required. In this review, we highlight the latest advances of therapeutic antibodies in MCL, alone or in combination with other strategies and agents, with a particular focus on the current challenges and future prospective.
Keywords: Cancer research, Immunology, Oncology, Pharmaceutical science
1. Introduction
In the past few decades, tremendous advances in treatment have been achieved in the field of hematological malignancies, including leukemias and lymphomas, especially non-Hodgkin's lymphomas (NHL) [1]. Mantle cell lymphoma (MCL) is an aggressive B-cell NHL characterized by the t(11; 14) (q13; q32) translocation and increased CCND1 (also termed cyclin D1) expression. The t(11; 14) (q13; q32) translocation leads to the constitutive overexpression of CCND1, which binds to cyclin-dependent kinases 4 and 6 (CDK4/6) and results in cell cycle disorder [2]. However, a subset of patients with MCL lack the t(11; 14) translocation and CCND1 expression; remarkably, CCND1 overexpression in mouse models is insufficient to induce spontaneous tumors [3], suggesting that other genetic or epigenetic events, signaling pathways, and microenvironmental alterations might be required for the initiation and progression of MCL [4, 5, 6, 7].
Due to the complex pathogenesis of MCL, there is no generally accepted standard of care specific for MCL patients at the initial stage, nor in the relapsed/refractory setting [8]. Although there are several chemotherapeutic backbones can be used in MCL, such as CHOP (cyclophosphamide, doxorubicin, vincristine, and prednisolone) and Hyper-CVAD (cyclophosphamide, vincristine, doxorubicin, and dexamethasone), and many novel agents including bortezomib, ibrutinib, and lenalidomide have been approved for use, MCL remains an incurable subtype of NHL with the poorest long-term outcome [9]. Chemoresistance is one of the major explanations for poor prognosis of MCL. Thus, there is a pressing need to develop new targeted therapeutics and precision medicine for MCL, and in particular, to integrate new therapies into rational combinations to overcome drug resistance in MCL.
Monoclonal antibodies (mAbs) are gaining an increasing role in the therapy of blood cancer, especially when incorporated into other modalities. Being a rare subtype of NHL, MCL has seldom been specifically discussed in terms of the use of therapeutic antibodies. The functions and mechanisms of mAbs as well as the application of mAbs in other B-cell malignancies rather than MCL have been extensively reviewed [10, 11, 12, 13, 14], and they will not be discussed in depth here. In this article, we will go through the current and evolving therapeutic antibodies, alone or combined with other mAbs, chemotherapies, or novel agents in MCL exclusively unless otherwise specified, with a particular focus on the challenges and future directions.
2. Main text
2.1. Anti-CD20 mAbs in MCL
2.1.1. Rituximab (RTX)
The most important step in improving the outcome in NHL is the use of the type I chimeric anti-CD20 mAb RTX, a human/murine chimeric monoclonal antibody which has been clearly recognized as the most extensively employed treatment for NHL by inducing cytotoxicity via antibody-dependent cell-mediated cytotoxicity (ADCC), complement activation, and apoptosis (Fig. 1 and Table 1) [15, 16, 17]. RTX specifically targets the CD20 molecule on the surface of mature B cells and B-cell lymphomas. In MCL, RTX has moderate single-agent efficacy [18]. In 2000, a European multicenter phase II study was conducted to assess the toxicity, overall response rate (ORR), and complete remission (CR) rate to RTX. Seventy-four patients with newly diagnosed MCL (MCL1, n = 34) and previously treated MCL (MCL2, n = 40) received RTX for four weeks via intravenous infusion. Upon treatment, 38% of MCL1 and 37% of MCL2 cases achieved ORR, and approximately 15% of patients achieved CR [19]. Similarly, a retrospective analysis from 81 evaluable MCL patients treated with RTX showed 37% of patients achieved ORR with 14% of patients achieving CR; however, the median time to disease progression (TTP) was only seven months, suggesting that single-agent RTX is active in MCL but fails to cure patients [20].
Fig. 1.
Mechanisms of mAbs-mediated cell death in MCL. RTX is a human/murine chimeric anti-CD20 mAb, which leads to cell death via at least four mechanisms. Compared to RTX, humanized anti-CD20 mAb obinutuzumab and fully human anti-CD20 mAb ofatumumab have similar mechanisms, but each has its own distinct properties (not shown). To simplify the diagram, RTX is taken as an example to illustrate the mechanisms: a) RTX links natural killer (NK) cells to MCL cells through FcγR, resulting in ADCC by releasing cytolytic granules; b) the crosslinking of RTX and the CD20 antigen activates the complement cascade, forming the membrane attack complex (MAC) and promoting cell lysis via CDC; c) RTX that binds to CD20 can be recognized by the complement receptors (CRs) and FcγR expressed on macrophages, leading to antibody-dependent cellular phagocytosis (ADCP); d) multiple binding of RTX to CD20 can cause direct apoptosis. Radiolabeled anti-CD20 mAbs, 131I-tositumomab and 90Y-ibritumomab tiuxetan, generate dsDNA breaks by particle emissions. Both humanized anti-CD37 otlertuzumab and anti-CD74 milatuzumab induce direct apoptosis in MCL. The pro-death activity of milatuzumab is enhanced when combined with an autophagy inhibitor FTY720 by preventing degradation of CD74 in an autophagy-dependent manner. Blinatumomab is a murine bsAb targeting both the CD19 and CD3 antigens. Blinatumomab links CD3+ polyclonal T cells to CD19+ target cells, which activates T cells followed by serial cell lysis of CD19+ MCL cells and induces a robust T-cell proliferation as well as a transient release of cytokines.
Table 1.
Therapeutic mAbs in MCL.
| mAbs | Target | Origin | Isotype and format | Mechanisms |
|---|---|---|---|---|
| RTX | CD20 | Murine-human chimeric | Human IgG1κ | ADCC, CDC, ADCP, direct apoptosis |
| Ofatumumab | CD20 | Human | Human IgG1κ | ADCC, potent CDC, ADCP, direct apoptosis |
| Obinutuzumab | CD20 | Humanized | Human IgG1 | Enhanced ADCC, reduced CDC, ADCP, potent direct apoptosis |
| 131I-tositumomab | CD20 | Murine | 131I-radiolabled IgG2a | Double-strand DNA breaks caused by beta and gamma emissions |
| 90Y-ibritumomab tiuxetan | CD20 | Murine | 90Y-radiolabled IgG1κ | Double-strand DNA breaks caused by beta emission |
| Otlertuzumab | CD37 | Humanized | Human IgG1 | Direct apoptosis |
| Milatuzumab | CD74 | Humanized | Human IgG1 | Enhanced pro-death activity when combined with FTY720 by preventing degradation of CD74 in an autophagy-dependent manner |
| Galiximab | CD80 | Primate-human chimeric | Human IgG1λ | ADCC and apoptosis in NHL, but unknown in MCL |
| Cirmtuzumab | ROR1 | Humanized | Human IgG1 | Inhibit Wnt5a-enhanced cell proliferation |
| Blinatumomab | CD19/CD3 | Murine, bispecific | scFv-scFv | Robust T-cell activation to exert cytotoxic activity on target cells |
| HexAbs | CD20/CD74 | Bispecific | DNL conjugation | Direct potent cytotoxicity |
scFv, single-chain variable fragment.
Compared to its single-agent activity, RTX combined with chemotherapeutic regimen CHOP achieved 96% ORR including 48% CR in newly diagnosed MCL, however 70% of patients finally occurred relapse or developed progressive disease, implying that RTX with conventional chemotherapy may transiently clear tumor cells but not translate into prolonged progression-free survival (PFS) in MCL [21]. In 2004, therapeutic efficacy were reported with fludarabine, cyclophosphamide and mitoxantrone (FCM), alone or combined with RTX (R-FCM) in patients with relapsed/refractory MCL from a prospective randomized multicenter phase III trial. Of the 48 evaluable MCL patients (24 randomized for FCM, 24 randomized for R-FCM), R-FCM revealed a substantial benefit with 58% of ORR (29% CR, 29% partial remission [PR]) compared with 46% for FCM alone (0% CR, 46% PR); in addition, superior median PFS (p = 0·038) and overall survival (OS, p = 0·003) were obtained in the R-FCM arm compared with FCM alone, and no differences were found in clinically relevant side effects in both study arms [22]. Notably, RTX in combination with chemotherapy as either first-line therapy or maintenance therapy could particularly improve OS in elderly patients with MCL [23, 24]. In younger MCL patients, RTX maintenance after autologous stem-cell transplantation (ASCT) was also shown to improve PFS, event-free survival (EFS), and OS in a randomized phase III trial [25]. Very recently, a phase II study by Japan Clinical Oncology Group-Lymphoma Study Group (JCOG-LSG) showed high efficacy and acceptable toxicity of R-High-CHOP/CHASER (cyclophosphamide, high-dose cytarabine, dexamethasone, etoposide, and rituximab)/LEED (melphalan, cyclophosphamide, etoposide, and dexamethasone) plus ASCT in younger patients with untreated advanced MCL, providing a potential standard treatment option for newly diagnosed younger MCL patients [26]. More RTX-based chemotherapies in MCL have been well documented [8, 17].
In addition to chemotherapies, newer agents in combination with RTX have also been investigated. In a phase I/II clinical trial, combining RTX with lenalidomide, an oral immunomodulator with anti-neoplastic and anti-proliferative effects against MCL [27], resulted in an ORR of 57% (36% CR, 20% PR) with a median PFS of 11·1 months [28]. The efficacy of this combination appears even higher as an initial therapy for patients with previously untreated MCL [29]. Of note, RTX plus lenalidomide enhances efficacy over what has been shown with monotherapy and improves outcomes in the RTX-resistant patients [30, 31]. In addition to lenalidomide, bortezomib, a novel proteasome inhibitor approved in the U.S for the treatment of patients with MCL [32], has been incorporated into many regimens. As a part of front-line therapy, the combination of bortezomib with R-CHOP (RTX and CHOP) [33] or R-Hyper-CVAD (RTX and Hyper-CVAD) [34] obtains a striking advance over the original regimens with less toxicity. Ibrutinib, an oral covalent inhibitor of Bruton's tyrosine kinase (BTK), is able to irreversibly inactivate the B-cell receptor signaling pathway [35]. In a single-center open-label phase II trial, ibrutinib combined with RTX is active and well-tolerated in relapsed/refractory MCL patients with 88% of ORR (44% CR, 44% PR) [36]. Interestingly, the objective response was 100% in patients with Ki-67 < 50%, whereas worse treatment outcomes were observed in patients with higher Ki-67 levels (≥50%), suggesting that Ki-67 might serve as a predictor for this combination therapy in MCL [36]. Ibrutinib is also well tolerated when added to R-CHOP in a non-randomized phase Ib study [37]. Further combination of ibrutinib with RTX and bendamustine (R-bendamustine) achieved 94% ORR (76% CR) in newly diagnosed MCL patients [38] compared with 68% for single agent ibrutinib (21% CR) [39] and 75%–92% for R-bendamustine (41%–50% CR) in MCL [40, 41], although longer follow-ups and more clinical data such as the PFS are warranted for further evaluation. The clinical data of RTX-based studies are summarized in Table 2.
Table 2.
Monoclonal antibody-based therapies in MCL.
| Therapeutic regimen | Study features | Number of MCL patients | ORR (CR) (%) | PFS, TTP, EFS or other parameters (months) | Median OS (months) | Ref. |
|---|---|---|---|---|---|---|
| Single-agent RTX (multicenter) | Phase II | 74 (MCL1, n = 34; MCL2, n = 40) | 38 in MCL1 and 37 in MCL2 (15) | Median response duration: 1·2 yr | – | [19] |
| Single-agent RTX (multicenter) | Retrospective | 87 (MCL1, n = 37; MCL2, n = 50) | 37 (14) | TTP: 7 | – | [20] |
| R-CHOP | Phase II | 40 | 96 (48) | Median PFS: 16·6 | – | [21] |
| R-FCM vs FCM | Phase III | 48 | 58 (29) vs 46 (0) | Median PFS: 8 vs 4 | NR vs 11; 2-yr OS: 65% vs 35% | [22] |
| RTX + chemotherapy vs chemotherapy | – | 638 | – | – | 37 vs 27 | [23] |
| RTX + ASCT vs ASCT | Phase III | 240 (120/group) | – | 4-yr PFS: 83% vs 64%; 4-yr EFS: 79% vs 61% |
4-yr OS: 89% vs 80% | [25] |
| R-High-CHOP/CHASER/LEED + ASCT | Phase II | 45 (n = 35 for patients who completed ASCT) | 96 (82) (n = 45); 100 (91) (n = 35) |
5-yr PFS: 52% (n = 45) 5-yr PFS: 55% (n = 35) |
5-yr OS: 71% (n = 45); 5-yr OS: 72% (n = 35) |
[26] |
| RTX + lenalidomide | Phase I/II | 52 | 57 (36) | Median PFS: 11·1 | 24·3 | [28] |
| RTX + lenalidomide (multicenter) | Phase II | 38 | 92 (64) | Median PFS: NR; 2-yr PFS: 85% | 2-yr OS: 97% | [29] |
| RTX + lenalidomide | Phase II | 11 | 55 (55) | Median PFS: 24·4 | – | [31] |
| R-CHOP + bortezomib | Phase I/II | 36 | 91 (72) (n = 32); 81 (64) (n = 36) |
Median PFS: 23; 2-yr PFS: 44% (n = 36) | NR; 2-yr OS: 86% | [33] |
| RTX + ibrutinib | Phase II | 50 | 88 (44) | Median PFS: NR | NR | [36] |
| R-bendamustine + ibrutinib | Phase I/Ib | 17 | 94 (76) | Median PFS: NR | NR | [38] |
| Single-agent ofatumumab | Phase II | 12 | 8·3 (0) | – | 11·2 | [46] |
| Single-agent obinutuzumab | Phase II | 15 | 27 (13) | – | – | [56] |
| 131I-tositumomab + CHOP | – | 25 | 86 (67) | – | 2-yr OS: 92% | [59] |
| R-CHOP + 90Y-ibritumomab tiuxetan | Phase II | 53 | 88 (55) | Median PFS: 31 | 2-yr OS: 90% | |
| Single-agent 90Y-ibritumomab tiuxetan | Phase II | 34 | 31 (15) (n = 32) | Median EFS: 6 | 21 | [60] |
| 90Y-ibritumomab tiuxetan + bortezomib | Phase I | 5 | 80 (60) | – | NR | [61] |
| Single-agent otlertuzumab | Expansion of phase I | 4 | 0 | – | – | [65] |
| Single-agent blinatumomab (multicenter) | Phase I | 6 (15, 30 μg/m2/day); 2 (60 μg/m2/day) |
16·7 (16·7) (15, 30 μg/m2/day); 100 (50) (60 μg/m2/day) |
– | – | [94] |
| Single-agent blinatumomab | Phase I | 7 (60 μg/m2/day) | 71 (43) (60 μg/m2/day) |
– | – | [95] |
MCL1, newly diagnosed MCL; MCL2, previously treated MCL; NR, not reached.
2.1.2. Ofatumumab
Despite improved outcomes with RTX, a significant proportion of patients still experience disease relapse or refractory situations. In this context, alternative therapeutic strategies that can overcome resistance with more efficiency, such as second generation anti-CD20 mAbs, have been developed [1]. Ofatumumab is one of the second generation type I anti-CD20 mAbs, which was generated from human immunoglobulin transgenic mice (Table 1) [42, 43]. Compared to RTX, the fully human mAb ofatumumab binds to a distinct epitope of the CD20 molecule, which consists of the smaller extracellular loop and the N-terminal region of the large extracellular loop [43], much closer to the cell membrane. These favorable properties lead to an outstanding potency in complement-dependent cytotoxicity (CDC) and a lower off-rate to CD20 than RTX [42, 44]. In MCL cell lines, ofatumumab exhibits more potent CDC activity compared with RTX, including in cell lines with resistance to RTX; in a murine model of MCL, ofatumumab also delays tumor progression and prolongs survival [45]. Nevertheless, an open-label phase II single-arm study of ofatumumab monotherapy for relapsed/refractory MCL was carried out in three centers from the UK [46]. Of the 12 patients enrolled in the trial, the ORR was 8·3% with a median OS of 11·2 month; only one of the 12 patients achieved PR and six patients achieved stable disease, whereas the five remaining patients had progressive disease (Table 2) [46]. Of note, ofatumumab shows significant adverse event of fatigue (75% patients affected) that is much higher than other types of lymphomas [47]. Based on this study, the response rate of single-agent ofatumumab in relapsed/refractory MCL patients is low, although another study reported a complete remission after treatment with single-agent ofatumumab in a patient with high-risk leukemic MCL [48]. A more intensive and prolonged exposure to ofatumumab, or combination of ofatumumab with other anti-CD20 mAbs or chemotherapies might increase the potential of ofatumumab in MCL [46].
2.1.3. Obinutuzumab
Unlike type I anti-CD20 mAbs (e.g., RTX, ofatumumab), obinutuzumab is a novel humanized and glycoengineered type II anti-CD20 antibody, which induces enhanced ADCC and stronger direct cell death activity but does not localize mAb-CD20 antigen complexes into lipid rafts, resulting in lower levels of CDC (Table 1) [49]. Despite recognizing an overlapping epitope of the CD20 extracellular domain, obinutuzumab targets CD20 in a completely different orientation than type I antibodies [50], and exhibits superior efficacy over RTX and ofatumumab [51], especially in RTX-resistant cases [49]. One molecular mechanism is Fc-gamma receptors IIb (FcγRIIb), a key molecular expressed on B cells that can promote the internalization of RTX and result in reduced efficacy of RTX [52]. This finding implicates that type II anti-CD20 mAbs may enhance immune effector function by circumventing the limitations of internalization, although it is unclear why type II anti-CD20 mAbs tend not to internalize and do not engage and activate FcγRIIb [52].
In MCL, one study showed that drug resistance can be overcome with obinutuzumab which counteracted Bcl-xL induction through NF-kB inhibition; by contrast, RTX was not able to overcome drug resistance [53]. In another study, MCL cell lines Granta-519, Jeko-1, Rec-1, and Z-138 were treated with RTX or obinutuzumab, alone or in combination [54]. Obinutuzumab induced a higher reduction in cell viability in each MCL cell line compared to RTX, whereas combination experiments showed the competitive binding of these two mAbs due to overlapping epitopes on the target cells, leading to a lower cytotoxicity than obinutuzumab alone. Of interest, uniquely deregulated genes were induced after RTX and obinutuzumab mono-treatment, respectively, providing more rational strategies for new combination experiments in MCL [54]. In a phase I study (GAUGUIN), 43% of 21 heavily pretreated patients with NHL achieved disease response to 8×21-day cycles of obinutuzumab [55]; Later that year, the efficacy and safety of obinutuzumab were further explored by the same research group in one randomized phase II trial in relapsed/refractory MCL patients, in which, obinutuzumab achieved early steady-state concentration and clinical activity with an acceptable safety profile (Table 2) [56]. Ongoing and future trials will provide more evidence regarding the use of obinutuzumab in MCL, and other type II anti-CD20 mAbs are also anticipated.
2.1.4. Armed anti-CD20 mAbs
The addition of a radionuclide to monoclonal antibodies was thought to be a rational approach to achieve extra cytotoxic effects, particularly due to the inherent radio-sensitivity of most lymphomas including MCL. This modification, also termed radioimmunotherapy (RIT), has been reported as a safe and effective therapeutic option for NHL.
Two anti-CD20 RIT antibodies, iodine-131 (131I)-tositumomab [57] and yttrium-90 (90Y)-ibritumomab tiuxetan [58], have been approved by the Food and Drug Administration (FDA). Both agents have been reported for the treatment of relapsed/refractory CD20+ lymphomas. The radionuclides delivered by these two antibodies are decayed by releasing beta particles, thereby causing double-strand DNA (dsDNA) breaks in tumor cells (Fig. 1 and Table 1) [59]. In relapsed/refractory MCL, single-agent 90Y-ibritumomab tiuxetan earned an ORR of 31% (15% CR) and a median EFS of 28 months for patients who achieved PR or CR [60]. Sequential RIT with 131I-tositumomab followed by CHOP consolidation in MCL patients could induce molecular remission with 86% of ORR (67% CR) and 92% of OS at two years [59]. Similarly, 90Y-ibritumomab tiuxetan consolidation after R-CHOP chemotherapy in newly diagnosed MCL also obtained an ORR of 88%, and CR was dramatically elevated from 13% upon R-CHOP to 55% after RIT consolidation [59]. In addition to conventional chemotherapeutic regimens, RIT plus bortezomib were also evaluated in a phase I clinical trial: 90Y-ibritumomab tiuxetan preceded by bortezomib as a radiosensitizer is a safe and well-tolerated combination therapy with an ORR of 80% in relapsed/refractory MCL [61]. Given the radiosensitive feature of MCL, RIT allows for a more targeted therapeutic option in the treatment of MCL, both in the frontline and at relapse [62]. The clinical details of radioimmunotherapy in MCL are shown in Table 2.
2.2. Other potential mAbs in MCL
CD37 is a heavily glycosylated cell surface tetraspanin expressed on normal and malignant B cells [63, 64]. Due to its absence or weak expression on blood cells except B cells, CD37 is considered as a lineage-specific marker of human B cells, representing a key therapeutic target for B-cell malignancies [65]. In MCL, hypomethylated and overexpressed CD37 gene is revealed as a novel target for drug development from a genome-wide DNA methylation analysis, suggesting that distinct epigenetic changes could be targeted for therapeutic benefit in MCL [66].
Otlertuzumab is a humanized anti-CD37 protein therapeutic, and it triggers cell apoptosis directly by up-regulation of a proapoptotic protein BCL2 like 11 (BCL2L11, also termed BIM) in B-cell malignancies (Fig. 1 and Table 1) [67]. In a SCID mouse model of leukemia/lymphoma, significant in vivo therapeutic efficacy of otlertuzumab is revealed [68]. More importantly, otlertuzumab could offer an alternative therapeutic regimen when CD20 is blocked or even lost on the targeted B cells [69]. Therefore, it is unsurprised that otlertuzumab in combination with RTX or other chemotherapeutics leads to an enhanced anti-tumor activity in NHL models [65]. Nonetheless, the use of otlertuzumab in MCL has been rarely reported. In 2015, the clinical activity of otlertuzumab in patients with advanced MCL was firstly evaluated [65]. Among four patients with MCL, all had received prior RTX therapy and chemotherapy; in fact, otlertuzumab activity as a single agent in such a heavily pretreated population was not satisfactory in MCL, because none of the four MCL patients had a response (Table 2) [65]. Given these results, further studies with otlertuzumab in combination with RTX or other chemotherapeutic agents should be carried out in MCL.
Another potential target is CD74 antigen, which is an integral membrane protein with diverse functions. Enhanced expression of CD74 has been implicated in malignant B cell growth and survival, making it a potential target for immunotherapy [70]. Milatuzumab is a humanized mAb specific for CD74 (Table 1) [71]. Unlike RTX, milatuzumab does not induce ADCC or CDC [72], implying that milatuzumab targets distinct cell death signaling pathways via different cellular mechanisms. Thus, the preclinical combination of RTX and milatuzumab shows both in vitro and in vivo activity in MCL, and the same is true when incorporating milatuzumab into either ofatumumab or veltuzumab (anti-CD20 IgG) treatment [73]. Interestingly, the same research group also found FTY720, an immunosuppressive drug that serves as an autophagy-inhibiting agent in MCL cell lines; after treatment, FTY720 enhanced the pro-death activity of milatuzumab by preventing the degradation of CD74 in an autophagy-dependent manner (Fig. 1) [74, 75]. Afterwards, this group further reported OSU-2S, a novel non-immunosuppressive FTY720 derivative that could increase the surface expression of CD74. Therefore, OSU-2S in combination with milatuzumab could enhance cytotoxicity in MCL cells [76].
In addition to CD37 and CD74, CD80 is a member of the B7 family of immune coregulatory proteins that mediate both immune activation and suppression. CD80 has demonstrated a key role in regulating immune inhibitory processes [77], and in particular, it has been identified as a therapeutic target in NHL based on limited immunohistochemical studies [78, 79]. CD80 expression has been evaluated by flow cytometry analysis using primary lymphoma cells from patients with NHL, among which, 75% of MCL cases are CD80 positive [80]. Galiximab, a primate-human chimeric anti-CD80 antibody, has shown safety and efficacy in patients with relapsed/refractory follicular lymphoma (FL) (Table 1) [81]. Despite no report of galiximab in MCL yet, targeting CD80 could probably provide a clue in the treatment of MCL in near future.
Other than CD (cluster differentiation) molecules, a receptor tyrosine kinase-like orphan receptor 1 (ROR1) was identified as a dominant cell surface marker in chronic lymphoblastic leukemia (CLL) from two independent gene expression studies [82, 83]. ROR1 functions as a receptor for Wnt5a which induces noncanonical Wnt signaling, leading to activation of Rac1 and enhanced proliferation of CLL cells [84]. Intriguingly, ROR1 expression is not restricted to CLL, but is more prevalent in NHLs, especially in MCL where ROR1 is expressed intensely [85]. In this context, a humanized anti-ROR1 mAb cirmtuzumab becomes a potential target against MCL. Recently, one study revealed that MCL proliferation can be inhibited by cirmtuzumab via a ROR1-dependent pathway which, however cannot be blocked by ibrutinib, suggesting that cirmtuzumab and ibrutinib may play complementary roles in the treatment of MCL patients [86]. The synergistic anti-tumor effects of cirmtuzumab and ibrutinib has been proved last year in CLL cells [87], and the combination of cirmtuzumab with ibrutinib in MCL need further evaluation.
2.3. Bispecific antibodies (bsAb) in MCL
Combination of antibody (Ab) therapy can be achieved by a single bsAb to avoid administrating two different Abs sequentially, which is time-consuming, inconvenient, and expensive. Consequently, developing bsAbs is emerging as a new part of Ab therapies by generating various constructs differing in design, structure, and antigen (Ag)-binding properties. A desirable bsAb may serve to 1) recruit effector cells or effector molecules to target malignant cells, 2) improve the target specificity by simultaneous ligation two different Ags expressed on the same cell, or 3) involve more than one signaling pathway or mechanism controlling cell proliferation or survival. These features enable bsAbs a more potent function by overcoming single-mAb resistance and enhancing functional affinity, hence achieving a higher potency.
2.3.1. CD19/CD3 bsAb
Blinatumomab is a CD19/CD3 bispecific T cell-engaging antibody targeting both the CD3ε subunit of the T-cell receptor (TCR) complex and the CD19, a B-cell Ag which is expressed on malignant B cells across all NHL subtypes (Table 1) [88, 89]. Regular antibodies cannot directly recruit T cells due to the lack of Fc-gamma receptors (FcγR) on these cells, whereas blinatumomab links CD3+ polyclonal T cells to CD19+ B cells, which activates T cells followed by serial lysis of CD19+ target cells via granzymes and perforin [89, 90], and induces a robust T-cell expansion as well as a transient release of cytokines (Fig. 1) [91, 92, 93]. In 2008, the clinical activity and safety of single-agent blinatumomab was assessed in NHLs including MCL [94]. Partial and complete tumor regressions were first observed (4/19) at a dose of 15–30 μg/m2/day, and all patients (7/7) got a tumor regression at a dose of 60 μg/m2/day; notably, blinatumomab doses of 15 μg/m2/day and higher led to clearance of tumor cells not only in peripheral blood, lymph node lesions, and spleen, but also in bone marrow and infiltrated liver (Table 2) [94]. In a following phase I dose-escalation study, a total of 24 patients with relapsed/refractory MCL were enrolled; the ORR was 71% (n = 7, 3 CR/unconfirmed CR [CRU] and 2 PR) with blinatumomab monotherapy at a dose of 60 μg/m2/day, which was established as the maximum tolerated dose (MTD) for administration and was finally selected as the target dose for efficacy, although one patient did not respond to retreatment at 60 μg/m2/day and experienced a relapse (Table 2) [95]. Future developments including the potential combination of blinatumomab with regular chemotherapies or other agents, and the extension of alternative routes of administration need to be further addressed and optimized [89].
2.3.2. CD20/CD74 bsAb
Owing to the fact that RTX and milatuzumab target different antigens and signaling pathways, one group explored a preclinical evaluation of combination strategy in MCL: targeting both CD20 and CD74 led to rapid cell death in MCL cell lines and primary tumor cells compared with either parental Ab alone [73]. Later that year, the same team successfully generated novel bispecific hexavalent Abs (HexAbs) from veltuzumab and milatuzumab using a Dock-and-Lock (DNL) method; surprisingly, the anti-CD20/CD74 bispecific HexAbs resulted in homotypic adhesion and triggered multiple intracellular events in MCL cell lines and primary MCL cells including production of reactive oxygen species (ROS), rapid and sustained activation of ERKs and JNK-MAPKs, deactivation of the PI3K/Akt signaling pathway, and actin reorganization, etc. [96] This study indicates that such bsAb constructs are more potent than either parental mAb or even combination of these mAbs, and is likely to provide new immunotherapeutic strategies for the other CD20+/CD74+ malignancies.
2.4. Current challenges and future prospective
Over the past decades, mAbs have proven to be incredibly valuable as cancer therapeutics and have made substantial achievement in clinical outcome. Although RTX has been incorporated routinely into regimens and has made an important impact on NHL, there has been no significant therapeutic improvement for patients with MCL for many years [17].
RTX resistance is a great challenge in the treatment of MCL. The mechanisms underlying resistance are still unclear. Reduction or loss of the CD20 hallmark from the lymphoma cells has been reported as a major contributor to unresponsiveness [97, 98, 99, 100, 101]. One mechanism of CD20 loss is the “shaving reaction”, in which RTX-CD20 complex is cleared from MCL cells by monocytes in the circulation mediated by FcγR [102]. Another mechanism is the CD20 internalization modulated by type I anti-CD20 mAbs, shortening mAb half-life via a rapid degradation of CD20/mAb complex in lysosomes [103]. FcγRIIb is further revealed as a key participant to promote CD20 internalization [52]. Importantly, these findings probably explain the different sensitivity of B-cell malignancies to RTX. For example, CD20 internalization is the most evident in MCL samples which express higher FcγRIIb [52, 103], consistent with the relatively poor response of MCL to RTX treatment; by contrast, significantly lower internalization and FcγRIIb expression are observed in FL and diffuse large B-cell lymphoma (DLBCL) samples which are far more resistant to CD20 loss [52, 103]. Other possible resistance mechanisms include alterations of tumor cells survival pathways (ERKs, p38 MAPK, NF-κB, etc.), leading to failure of RTX to target these signaling [104]; defective RTX-induced redistribution of CD20 in lipid rafts, failing to evoke CDC, calcium influx and apoptosis [105, 106, 107]; FcγR polymorphisms that can increase RTX affinity in FL and can predict the outcome in some studies of FL and DLBCL [108, 109, 110], but it is independent of the RTX efficacy in MCL [111]. Despite these potential mechanisms for disparity in efficacy among different NHL subtypes, MCL continues to be a therapeutic challenge due to its unclear pathogenesis and very limited research.
Owing to poor response to RTX, advances in the design of other anti-CD20 mAbs, such as ofatumumab and obinutuzumab, have offered alternative options for the treatment of MCL. However, it is hard to make a clear comparison between RTX and other types of anti-CD20 mAbs, because most of patients have already received prior RTX therapy. Therefore, well-designed and -executed clinical trials are needed to determine the true clinical benefit of each mAb and its long-term safety. Apart from CD20, other Ags including CD37, CD74, CD80, and ROR1 also exhibit great potentials in MCL, although very limited studies are reported.
Step forward, multiple newer drugs have incorporated into the standard regimens. In addition to bortezomib, ibrutinib, and lenalidomide, agents that are being explored or under investigation comprise PI3K inhibitor idelalisib, BCL2 inhibitors, HDAC inhibitors, and CDK4/6 inhibitors etc., and we are still in a “watching and waiting” stage. It is beyond the scope of this article to discuss these agents in detail, however we can anticipate that over the next decades, we will be very likely to embrace a brand-new chemo-free model of treatment with mAbs and target agents in MCL. Nevertheless, the management of relapsed/refractory MCL with high tumor burden still requires a long duration to be fully realized, and chemotherapy-free regiments must be cautiously considered and performed.
It is noteworthy that most current reports focus on either basic research of mAbs engineering or clinical trials regarding the use of mAbs in MCL, and both are seriously disconnected to each other. Remarkably, the mechanism of mAb action rather than therapeutic efficacy in MCL patients are lacking, resulting in many unaddressed questions, for example, how multiple agents work together and interact in the human body? Why they do not work in some selected MCL patients? Compared to other types of NHLs, what is the unique mechanisms in MCL underlying the resistance to mAbs? How to stimulate effector cells appropriately without an overshooting immune response upon the treatment of bsAbs? It is necessary to go deeper into the research, not solely to assess clinical efficacy of mAbs, but also to figure out why or why not mAbs work, how they work, and which mechanism is to be favored when adding adjuvants, thereby providing evidence-based guidelines for treatment.
Another major challenge is how to collect solid clinical data with very limited population of MCL patients. Few studies have specifically targeted MCL, instead, most scientific and clinical data are along with other types of NHL. It seems hard and insufficient to draw a conclusion with only a small number of MCL cases enrolled in clinical trials. Therefore, a multicenter study from several groups around the world is probably the best way to solve this problem. Furthermore, off-target effects of mAbs therapy is also a big issue that should not be underestimated. Despite high potency and favorable cytotoxicity obtained by mAbs, they are not toxicity-free for normal tissues and cells, and potential side effects might be caused, especially when incorporated into other strategies.
Last but not the least, mounting evidence from earlier clinical trials suggests that initial therapy is very likely the best time to incorporate novel agents in order to minimize drug resistance and improve overall outcome [112]. Thus newly designed clinical trials in untreated MCL patients instead of heavily pretreated patients are warranted.
3. Conclusions
Therapeutic mAbs alone or combined with conventional chemotherapies or newer agents provide alternative potentials for the treatment of MCL. Despite great progress of utilizing mAbs in MCL, there is still much leeway for improvement. A better understanding of the tumor-specific mechanisms and pharmacokinetics, tailoring new anti-CD20 mAbs, developing novel mAbs and antibody-drug conjugates (ADC), designing bsAbs with brand-new concept, and identifying predictors for therapeutic response are urgently needed and will offer new opportunities to make better therapeutic mAbs or make mAbs better. The advances even breakthroughs in the treatment of MCL in the future is worth looking forward to.
Declarations
Author contribution statement
All authors listed have significantly contributed to the development and the writing of this article.
Funding statement
This work was supported by the Fundamental Research Funds for the Central Universities (No. 3332018130) given to H.Z.; Key Project from Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College given to H.Z.; Yunnan Open-end Funds of Joint Laboratory (No. 2016ynvdklop01) given to M.S.
Competing interest statement
The authors declare no conflict of interest.
Additional information
No additional information is available for this paper.
References
- 1.Karlin L., Coiffier B. Ofatumumab in the treatment of non-Hodgkin's lymphomas. Expert Opin. Biol. Ther. 2015;15(7):1085–1091. doi: 10.1517/14712598.2015.1055241. [DOI] [PubMed] [Google Scholar]
- 2.Jares P., Colomer D., Campo E. Molecular pathogenesis of mantle cell lymphoma. J. Clin. Invest. 2012;122(10):3416–3423. doi: 10.1172/JCI61272. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Adams J.M., Harris A.W., Strasser A., Ogilvy S., Cory S. Transgenic models of lymphoid neoplasia and development of a pan-hematopoietic vector. Oncogene. 1999;18(38):5268–5277. doi: 10.1038/sj.onc.1202997. [DOI] [PubMed] [Google Scholar]
- 4.Salaverria I., Royo C., Carvajal-Cuenca A., Clot G., Navarro A., Valera A. CCND2 rearrangements are the most frequent genetic events in cyclin D1(-) mantle cell lymphoma. Blood. 2013;121(8):1394–1402. doi: 10.1182/blood-2012-08-452284. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Bea S., Valdes-Mas R., Navarro A., Salaverria I., Martin-Garcia D., Jares P. Landscape of somatic mutations and clonal evolution in mantle cell lymphoma. Proc. Natl. Acad. Sci. U. S. A. 2013;110(45):18250–18255. doi: 10.1073/pnas.1314608110. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Zhang H., Chen Z., Miranda R.N., Medeiros L.J., McCarty N. TG2 and NF-kappaB signaling coordinates the survival of mantle cell lymphoma cells via IL6-mediated autophagy. Cancer Res. 2016;76(21):6410–6423. doi: 10.1158/0008-5472.CAN-16-0595. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Zhang H., Chen Z., Miranda R.N., Medeiros L.J., McCarty N. Bifurcated BACH2 control coordinates mantle cell lymphoma survival and dispersal during hypoxia. Blood. 2017;130(6):763–776. doi: 10.1182/blood-2017-02-767293. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Campo E., Rule S. Mantle cell lymphoma: evolving management strategies. Blood. 2015;125(1):48–55. doi: 10.1182/blood-2014-05-521898. [DOI] [PubMed] [Google Scholar]
- 9.Lenz G., Dreyling M., Hoster E., Wormann B., Duhrsen U., Metzner B. Immunochemotherapy with rituximab and cyclophosphamide, doxorubicin, vincristine, and prednisone significantly improves response and time to treatment failure, but not long-term outcome in patients with previously untreated mantle cell lymphoma: results of a prospective randomized trial of the German Low Grade Lymphoma Study Group (GLSG) J. Clin. Oncol. 2005;23(9):1984–1992. doi: 10.1200/JCO.2005.08.133. [DOI] [PubMed] [Google Scholar]
- 10.Waldmann T.A. Monoclonal antibodies in diagnosis and therapy. Science. 1991;252(5013):1657–1662. doi: 10.1126/science.2047874. [DOI] [PubMed] [Google Scholar]
- 11.Grossbard M.L., Press O.W., Appelbaum F.R., Bernstein I.D., Nadler L.M. Monoclonal antibody-based therapies of leukemia and lymphoma. Blood. 1992;80(4):863–878. [PubMed] [Google Scholar]
- 12.Jaglowski S.M., Alinari L., Lapalombella R., Muthusamy N., Byrd J.C. The clinical application of monoclonal antibodies in chronic lymphocytic leukemia. Blood. 2010;116(19):3705–3714. doi: 10.1182/blood-2010-04-001230. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Jabbour E., O'Brien S., Ravandi F., Kantarjian H. Monoclonal antibodies in acute lymphoblastic leukemia. Blood. 2015;125(26):4010–4016. doi: 10.1182/blood-2014-08-596403. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Hoyos V., Borrello I. The immunotherapy era of myeloma: monoclonal antibodies, vaccines, and adoptive T-cell therapies. Blood. 2016;128(13):1679–1687. doi: 10.1182/blood-2016-05-636357. [DOI] [PubMed] [Google Scholar]
- 15.Coiffier B. Rituximab in diffuse large B-cell lymphoma. Clin. Adv. Hematol. Oncol. 2004;2(3):156–157. [PubMed] [Google Scholar]
- 16.Coiffier B. Rituximab therapy in malignant lymphoma. Oncogene. 2007;26(25):3603–3613. doi: 10.1038/sj.onc.1210376. [DOI] [PubMed] [Google Scholar]
- 17.Salles G., Barrett M., Foa R., Maurer J., O'Brien S., Valente N. Rituximab in B-cell hematologic malignancies: a review of 20 years of clinical experience. Adv. Ther. 2017;34(10):2232–2273. doi: 10.1007/s12325-017-0612-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Foran J.M. Antibody-based therapy of non-Hodgkin's lymphoma. Best Pract. Res. Clin. Haematol. 2002;15(3):449–465. doi: 10.1053/beha.2002.0222. [DOI] [PubMed] [Google Scholar]
- 19.Foran J.M., Rohatiner A.Z., Cunningham D., Popescu R.A., Solal-Celigny P., Ghielmini M. European phase II study of rituximab (chimeric anti-CD20 monoclonal antibody) for patients with newly diagnosed mantle-cell lymphoma and previously treated mantle-cell lymphoma, immunocytoma, and small B-cell lymphocytic lymphoma. J. Clin. Oncol. 2000;18(2):317–324. doi: 10.1200/JCO.2000.18.2.317. [DOI] [PubMed] [Google Scholar]
- 20.Foran J.M., Cunningham D., Coiffier B., Solal-Celigny P., Reyes F., Ghielmini M. Treatment of mantle-cell lymphoma with Rituximab (chimeric monoclonal anti-CD20 antibody): analysis of factors associated with response. Ann. Oncol. 2000;11(Suppl. 1):117–121. [PubMed] [Google Scholar]
- 21.Howard O.M., Gribben J.G., Neuberg D.S., Grossbard M., Poor C., Janicek M.J. Rituximab and CHOP induction therapy for newly diagnosed mantle-cell lymphoma: molecular complete responses are not predictive of progression-free survival. J. Clin. Oncol. 2002;20(5):1288–1294. doi: 10.1200/JCO.2002.20.5.1288. [DOI] [PubMed] [Google Scholar]
- 22.Forstpointner R., Dreyling M., Repp R., Hermann S., Hanel A., Metzner B. The addition of rituximab to a combination of fludarabine, cyclophosphamide, mitoxantrone (FCM) significantly increases the response rate and prolongs survival as compared with FCM alone in patients with relapsed and refractory follicular and mantle cell lymphomas: results of a prospective randomized study of the German Low-Grade Lymphoma Study Group. Blood. 2004;104(10):3064–3071. doi: 10.1182/blood-2004-04-1323. [DOI] [PubMed] [Google Scholar]
- 23.Griffiths R., Mikhael J., Gleeson M., Danese M., Dreyling M. Addition of rituximab to chemotherapy alone as first-line therapy improves overall survival in elderly patients with mantle cell lymphoma. Blood. 2011;118(18):4808–4816. doi: 10.1182/blood-2011-04-348367. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Kluin-Nelemans H.C., Hoster E., Hermine O., Walewski J., Trneny M., Geisler C.H. Treatment of older patients with mantle-cell lymphoma. N. Engl. J. Med. 2012;367(6):520–531. doi: 10.1056/NEJMoa1200920. [DOI] [PubMed] [Google Scholar]
- 25.Le Gouill S., Thieblemont C., Oberic L., Moreau A., Bouabdallah K., Dartigeas C. Rituximab after autologous stem-cell transplantation in mantle-cell lymphoma. N. Engl. J. Med. 2017;377(13):1250–1260. doi: 10.1056/NEJMoa1701769. [DOI] [PubMed] [Google Scholar]
- 26.Ogura M., Yamamoto K., Morishima Y., Wakabayashi M., Tobinai K., Ando K. R-High-CHOP/CHASER/LEED with autologous stem cell transplantation in newly diagnosed mantle cell lymphoma: JCOG0406 STUDY. Cancer Sci. 2018;109(9):2830–2840. doi: 10.1111/cas.13719. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Qian Z., Zhang L., Cai Z., Sun L., Wang H., Yi Q. Lenalidomide synergizes with dexamethasone to induce growth arrest and apoptosis of mantle cell lymphoma cells in vitro and in vivo. Leuk. Res. 2011;35(3):380–386. doi: 10.1016/j.leukres.2010.09.027. [DOI] [PubMed] [Google Scholar]
- 28.Wang M., Fayad L., Wagner-Bartak N., Zhang L., Hagemeister F., Neelapu S.S. Lenalidomide in combination with rituximab for patients with relapsed or refractory mantle-cell lymphoma: a phase 1/2 clinical trial. Lancet Oncol. 2012;13(7):716–723. doi: 10.1016/S1470-2045(12)70200-0. [DOI] [PubMed] [Google Scholar]
- 29.Ruan J., Martin P., Shah B., Schuster S.J., Smith S.M., Furman R.R. Lenalidomide plus rituximab as initial treatment for mantle-cell lymphoma. N. Engl. J. Med. 2015;373(19):1835–1844. doi: 10.1056/NEJMoa1505237. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Ruan J., Shah B., Martin P., Schuster S.J. Clinical experience with lenalidomide alone or in combination with rituximab in indolent B-cell and mantle cell lymphomas. Ann. Oncol. 2016;27(7):1226–1234. doi: 10.1093/annonc/mdw158. [DOI] [PubMed] [Google Scholar]
- 31.Chong E.A., Ahmadi T., Aqui N.A., Svoboda J., Nasta S.D., Mato A.R. Combination of lenalidomide and rituximab overcomes rituximab resistance in patients with indolent B-cell and mantle cell lymphomas. Clin. Cancer Res. 2015;21(8):1835–1842. doi: 10.1158/1078-0432.CCR-14-2221. [DOI] [PubMed] [Google Scholar]
- 32.Goy A., Bernstein S.H., Kahl B.S., Djulbegovic B., Robertson M.J., de Vos S. Bortezomib in patients with relapsed or refractory mantle cell lymphoma: updated time-to-event analyses of the multicenter phase 2 PINNACLE study. Ann. Oncol. 2009;20(3):520–525. doi: 10.1093/annonc/mdn656. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Ruan J., Martin P., Furman R.R., Lee S.M., Cheung K., Vose J.M. Bortezomib plus CHOP-rituximab for previously untreated diffuse large B-cell lymphoma and mantle cell lymphoma. J. Clin. Oncol. 2011;29(6):690–697. doi: 10.1200/JCO.2010.31.1142. [DOI] [PubMed] [Google Scholar]
- 34.Romaguera J.E., Fayad L.E., McLaughlin P., Pro B., Rodriguez A., Wang M. Phase I trial of bortezomib in combination with rituximab-HyperCVAD alternating with rituximab, methotrexate and cytarabine for untreated aggressive mantle cell lymphoma. Br. J. Haematol. 2010;151(1):47–53. doi: 10.1111/j.1365-2141.2010.08315.x. [DOI] [PubMed] [Google Scholar]
- 35.Honigberg L.A., Smith A.M., Sirisawad M., Verner E., Loury D., Chang B. The Bruton tyrosine kinase inhibitor PCI-32765 blocks B-cell activation and is efficacious in models of autoimmune disease and B-cell malignancy. Proc. Natl. Acad. Sci. U. S. A. 2010;107(29):13075–13080. doi: 10.1073/pnas.1004594107. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Wang M.L., Lee H., Chuang H., Wagner-Bartak N., Hagemeister F., Westin J. Ibrutinib in combination with rituximab in relapsed or refractory mantle cell lymphoma: a single-centre, open-label, phase 2 trial. Lancet Oncol. 2016;17(1):48–56. doi: 10.1016/S1470-2045(15)00438-6. [DOI] [PubMed] [Google Scholar]
- 37.Younes A., Thieblemont C., Morschhauser F., Flinn I., Friedberg J.W., Amorim S. Combination of ibrutinib with rituximab, cyclophosphamide, doxorubicin, vincristine, and prednisone (R-CHOP) for treatment-naive patients with CD20-positive B-cell non-Hodgkin lymphoma: a non-randomised, phase 1b study. Lancet Oncol. 2014;15(9):1019–1026. doi: 10.1016/S1470-2045(14)70311-0. [DOI] [PubMed] [Google Scholar]
- 38.Maddocks K., Christian B., Jaglowski S., Flynn J., Jones J.A., Porcu P. A phase 1/1b study of rituximab, bendamustine, and ibrutinib in patients with untreated and relapsed/refractory non-Hodgkin lymphoma. Blood. 2015;125(2):242–248. doi: 10.1182/blood-2014-08-597914. [DOI] [PubMed] [Google Scholar]
- 39.Wang M.L., Rule S., Martin P., Goy A., Auer R., Kahl B.S. Targeting BTK with ibrutinib in relapsed or refractory mantle-cell lymphoma. N. Engl. J. Med. 2013;369(6):507–516. doi: 10.1056/NEJMoa1306220. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Rummel M.J., Al-Batran S.E., Kim S.Z., Welslau M., Hecker R., Kofahl-Krause D. Bendamustine plus rituximab is effective and has a favorable toxicity profile in the treatment of mantle cell and low-grade non-Hodgkin's lymphoma. J. Clin. Oncol. 2005;23(15):3383–3389. doi: 10.1200/JCO.2005.08.100. [DOI] [PubMed] [Google Scholar]
- 41.Robinson K.S., Williams M.E., van der Jagt R.H., Cohen P., Herst J.A., Tulpule A. Phase II multicenter study of bendamustine plus rituximab in patients with relapsed indolent B-cell and mantle cell non-Hodgkin's lymphoma. J. Clin. Oncol. 2008;26(27):4473–4479. doi: 10.1200/JCO.2008.17.0001. [DOI] [PubMed] [Google Scholar]
- 42.Teeling J.L., French R.R., Cragg M.S., van den Brakel J., Pluyter M., Huang H. Characterization of new human CD20 monoclonal antibodies with potent cytolytic activity against non-Hodgkin lymphomas. Blood. 2004;104(6):1793–1800. doi: 10.1182/blood-2004-01-0039. [DOI] [PubMed] [Google Scholar]
- 43.Teeling J.L., Mackus W.J., Wiegman L.J., van den Brakel J.H., Beers S.A., French R.R. The biological activity of human CD20 monoclonal antibodies is linked to unique epitopes on CD20. J. Immunol. 2006;177(1):362–371. doi: 10.4049/jimmunol.177.1.362. [DOI] [PubMed] [Google Scholar]
- 44.Cang S., Mukhi N., Wang K., Liu D. Novel CD20 monoclonal antibodies for lymphoma therapy. J. Hematol. Oncol. 2012;5:64. doi: 10.1186/1756-8722-5-64. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Barth M.J., Mavis C., Czuczman M.S., Hernandez-Ilizaliturri F.J. Ofatumumab exhibits enhanced in vitro and in vivo activity compared to rituximab in preclinical models of mantle cell lymphoma. Clin. Cancer Res. 2015;21(19):4391–4397. doi: 10.1158/1078-0432.CCR-15-0056. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Furtado M., Dyer M.J., Johnson R., Berrow M., Rule S. Ofatumumab monotherapy in relapsed/refractory mantle cell lymphoma – a phase II trial. Br. J. Haematol. 2014;165(4):575–578. doi: 10.1111/bjh.12769. [DOI] [PubMed] [Google Scholar]
- 47.Czuczman M.S., Hess G., Gadeberg O.V., Pedersen L.M., Goldstein N., Gupta I. Chemoimmunotherapy with ofatumumab in combination with CHOP in previously untreated follicular lymphoma. Br. J. Haematol. 2012;157(4):438–445. doi: 10.1111/j.1365-2141.2012.09086.x. [DOI] [PubMed] [Google Scholar]
- 48.Hunstig F., Hammersen J., Kunert C., Petersen I., Merz H., Glaser A. Complete remission after treatment with single-agent ofatumumab in a patient with high-risk leukemic mantle-cell lymphoma. J. Clin. Oncol. 2013;31(19):e312–e315. doi: 10.1200/JCO.2012.45.9438. [DOI] [PubMed] [Google Scholar]
- 49.Illidge T., Klein C., Sehn L.H., Davies A., Salles G., Cartron G. Obinutuzumab in hematologic malignancies: lessons learned to date. Cancer Treat Rev. 2015;41(9):784–792. doi: 10.1016/j.ctrv.2015.07.003. [DOI] [PubMed] [Google Scholar]
- 50.Niederfellner G., Lammens A., Mundigl O., Georges G.J., Schaefer W., Schwaiger M. Epitope characterization and crystal structure of GA101 provide insights into the molecular basis for type I/II distinction of CD20 antibodies. Blood. 2011;118(2):358–367. doi: 10.1182/blood-2010-09-305847. [DOI] [PubMed] [Google Scholar]
- 51.Herter S., Herting F., Mundigl O., Waldhauer I., Weinzierl T., Fauti T. Preclinical activity of the type II CD20 antibody GA101 (obinutuzumab) compared with rituximab and ofatumumab in vitro and in xenograft models. Mol. Cancer Ther. 2013;12(10):2031–2042. doi: 10.1158/1535-7163.MCT-12-1182. [DOI] [PubMed] [Google Scholar]
- 52.Lim S.H., Vaughan A.T., Ashton-Key M., Williams E.L., Dixon S.V., Chan H.T. Fc gamma receptor IIb on target B cells promotes rituximab internalization and reduces clinical efficacy. Blood. 2011;118(9):2530–2540. doi: 10.1182/blood-2011-01-330357. [DOI] [PubMed] [Google Scholar]
- 53.Chiron D., Bellanger C., Papin A., Tessoulin B., Dousset C., Maiga S. Rational targeted therapies to overcome microenvironment-dependent expansion of mantle cell lymphoma. Blood. 2016;128(24):2808–2818. doi: 10.1182/blood-2016-06-720490. [DOI] [PubMed] [Google Scholar]
- 54.Heinrich D.A., Weinkauf M., Hutter G., Zimmermann Y., Jurinovic V., Hiddemann W. Differential regulation patterns of the anti-CD20 antibodies obinutuzumab and rituximab in mantle cell lymphoma. Br. J. Haematol. 2015;168(4):606–610. doi: 10.1111/bjh.13132. [DOI] [PubMed] [Google Scholar]
- 55.Salles G., Morschhauser F., Lamy T., Milpied N., Thieblemont C., Tilly H. Phase 1 study results of the type II glycoengineered humanized anti-CD20 monoclonal antibody obinutuzumab (GA101) in B-cell lymphoma patients. Blood. 2012;119(22):5126–5132. doi: 10.1182/blood-2012-01-404368. [DOI] [PubMed] [Google Scholar]
- 56.Morschhauser F.A., Cartron G., Thieblemont C., Solal-Celigny P., Haioun C., Bouabdallah R. Obinutuzumab (GA101) monotherapy in relapsed/refractory diffuse large b-cell lymphoma or mantle-cell lymphoma: results from the phase II GAUGUIN study. J. Clin. Oncol. 2013;31(23):2912–2919. doi: 10.1200/JCO.2012.46.9585. [DOI] [PubMed] [Google Scholar]
- 57.Kaminski M.S., Zelenetz A.D., Press O.W., Saleh M., Leonard J., Fehrenbacher L. Pivotal study of iodine I 131 tositumomab for chemotherapy-refractory low-grade or transformed low-grade B-cell non-Hodgkin's lymphomas. J. Clin. Oncol. 2001;19(19):3918–3928. doi: 10.1200/JCO.2001.19.19.3918. [DOI] [PubMed] [Google Scholar]
- 58.Witzig T.E., Gordon L.I., Cabanillas F., Czuczman M.S., Emmanouilides C., Joyce R. Randomized controlled trial of yttrium-90-labeled ibritumomab tiuxetan radioimmunotherapy versus rituximab immunotherapy for patients with relapsed or refractory low-grade, follicular, or transformed B-cell non-Hodgkin's lymphoma. J. Clin. Oncol. 2002;20(10):2453–2463. doi: 10.1200/JCO.2002.11.076. [DOI] [PubMed] [Google Scholar]
- 59.Tomblyn M. Radioimmunotherapy for B-cell non-hodgkin lymphomas. Cancer Contr. 2012;19(3):196–203. doi: 10.1177/107327481201900304. [DOI] [PubMed] [Google Scholar]
- 60.Wang M., Oki Y., Pro B., Romaguera J.E., Rodriguez M.A., Samaniego F. Phase II study of yttrium-90-ibritumomab tiuxetan in patients with relapsed or refractory mantle cell lymphoma. J. Clin. Oncol. 2009;27(31):5213–5218. doi: 10.1200/JCO.2009.21.8545. [DOI] [PubMed] [Google Scholar]
- 61.Beaven A.W., Shea T.C., Moore D.T., Feldman T., Ivanova A., Ferraro M. A phase I study evaluating ibritumomab tiuxetan (Zevalin(R)) in combination with bortezomib (Velcade(R)) in relapsed/refractory mantle cell and low grade B-cell non-Hodgkin lymphoma. Leuk. Lymphoma. 2012;53(2):254–258. doi: 10.3109/10428194.2011.608445. [DOI] [PubMed] [Google Scholar]
- 62.Skarbnik A.P., Smith M.R. Radioimmunotherapy in mantle cell lymphoma. Best Pract. Res. Clin. Haematol. 2012;25(2):201–210. doi: 10.1016/j.beha.2012.04.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Belov L., de la Vega O., dos Remedios C.G., Mulligan S.P., Christopherson R.I. Immunophenotyping of leukemias using a cluster of differentiation antibody microarray. Cancer Res. 2001;61(11):4483–4489. [PubMed] [Google Scholar]
- 64.Barrena S., Almeida J., Yunta M., Lopez A., Fernandez-Mosteirin N., Giralt M. Aberrant expression of tetraspanin molecules in B-cell chronic lymphoproliferative disorders and its correlation with normal B-cell maturation. Leukemia. 2005;19(8):1376–1383. doi: 10.1038/sj.leu.2403822. [DOI] [PubMed] [Google Scholar]
- 65.Pagel J.M., Spurgeon S.E., Byrd J.C., Awan F.T., Flinn I.W., Lanasa M.C. Otlertuzumab (TRU-016), an anti-CD37 monospecific ADAPTIR therapeutic protein, for relapsed or refractory NHL patients. Br. J. Haematol. 2015;168(1):38–45. doi: 10.1111/bjh.13099. [DOI] [PubMed] [Google Scholar]
- 66.Leshchenko V.V., Kuo P.Y., Shaknovich R., Yang D.T., Gellen T., Petrich A. Genomewide DNA methylation analysis reveals novel targets for drug development in mantle cell lymphoma. Blood. 2010;116(7):1025–1034. doi: 10.1182/blood-2009-12-257485. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Lapalombella R., Yeh Y.Y., Wang L., Ramanunni A., Rafiq S., Jha S. Tetraspanin CD37 directly mediates transduction of survival and apoptotic signals. Cancer Cell. 2012;21(5):694–708. doi: 10.1016/j.ccr.2012.03.040. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Zhao X., Lapalombella R., Joshi T., Cheney C., Gowda A., Hayden-Ledbetter M.S. Targeting CD37-positive lymphoid malignancies with a novel engineered small modular immunopharmaceutical. Blood. 2007;110(7):2569–2577. doi: 10.1182/blood-2006-12-062927. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Jilani I., O'Brien S., Manshuri T., Thomas D.A., Thomazy V.A., Imam M. Transient down-modulation of CD20 by rituximab in patients with chronic lymphocytic leukemia. Blood. 2003;102(10):3514–3520. doi: 10.1182/blood-2003-01-0055. [DOI] [PubMed] [Google Scholar]
- 70.Stein R., Mattes M.J., Cardillo T.M., Hansen H.J., Chang C.H., Burton J. CD74: a new candidate target for the immunotherapy of B-cell neoplasms. Clin. Cancer Res. 2007;13(18 Pt 2):5556s–5563s. doi: 10.1158/1078-0432.CCR-07-1167. [DOI] [PubMed] [Google Scholar]
- 71.Berkova Z., Tao R.H., Samaniego F. Milatuzumab – a promising new immunotherapeutic agent. Expert Opin. Investig. Drugs. 2010;19(1):141–149. doi: 10.1517/13543780903463854. [DOI] [PubMed] [Google Scholar]
- 72.Stein R., Qu Z., Cardillo T.M., Chen S., Rosario A., Horak I.D. Antiproliferative activity of a humanized anti-CD74 monoclonal antibody, hLL1, on B-cell malignancies. Blood. 2004;104(12):3705–3711. doi: 10.1182/blood-2004-03-0890. [DOI] [PubMed] [Google Scholar]
- 73.Alinari L., Yu B., Christian B.A., Yan F., Shin J., Lapalombella R. Combination anti-CD74 (milatuzumab) and anti-CD20 (rituximab) monoclonal antibody therapy has in vitro and in vivo activity in mantle cell lymphoma. Blood. 2011;117(17):4530–4541. doi: 10.1182/blood-2010-08-303354. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Alinari L., Mahoney E., Patton J., Zhang X., Huynh L., Earl C.T. FTY720 increases CD74 expression and sensitizes mantle cell lymphoma cells to milatuzumab-mediated cell death. Blood. 2011;118(26):6893–6903. doi: 10.1182/blood-2011-06-363879. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Alinari L., Baiocchi R.A., Praetorius-Ibba M. FTY720-induced blockage of autophagy enhances anticancer efficacy of milatuzumab in mantle cell lymphoma: is FTY720 the next autophagy-blocking agent in lymphoma treatment? Autophagy. 2012;8(3):416–417. doi: 10.4161/auto.19050. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Mani R., Chiang C.L., Frissora F.W., Yan R., Mo X., Baskar S. ROR1-targeted delivery of OSU-2S, a nonimmunosuppressive FTY720 derivative, exerts potent cytotoxicity in mantle-cell lymphoma in vitro and in vivo. Exp. Hematol. 2015;43(9):770–774 e2. doi: 10.1016/j.exphem.2015.04.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Curiel T.J., Wei S., Dong H., Alvarez X., Cheng P., Mottram P. Blockade of B7-H1 improves myeloid dendritic cell-mediated antitumor immunity. Nat. Med. 2003;9(5):562–567. doi: 10.1038/nm863. [DOI] [PubMed] [Google Scholar]
- 78.Dorfman D.M., Schultze J.L., Shahsafaei A., Michalak S., Gribben J.G., Freeman G.J. In vivo expression of B7-1 and B7-2 by follicular lymphoma cells can prevent induction of T-cell energy but is insufficient to induce significant T-cell proliferation. Blood. 1997;90(11):4297–4306. [PubMed] [Google Scholar]
- 79.Dogan A., Du M.Q., Aiello A., Diss T.C., Ye H.T., Pan L.X. Follicular lymphomas contain a clonally linked but phenotypically distinct neoplastic B-cell population in the interfollicular zone. Blood. 1998;91(12):4708–4714. [PubMed] [Google Scholar]
- 80.Dakappagari N., Ho S.N., Gascoyne R.D., Ranuio J., Weng A.P., Tangri S. CD80 (B7.1) is expressed on both malignant B cells and nonmalignant stromal cells in non-Hodgkin lymphoma. Cytom. B Clin. Cytom. 2012;82(2):112–119. doi: 10.1002/cyto.b.20631. [DOI] [PubMed] [Google Scholar]
- 81.Czuczman M.S., Thall A., Witzig T.E., Vose J.M., Younes A., Emmanouilides C. Phase I/II study of galiximab, an anti-CD80 antibody, for relapsed or refractory follicular lymphoma. J. Clin. Oncol. 2005;23(19):4390–4398. doi: 10.1200/JCO.2005.09.018. [DOI] [PubMed] [Google Scholar]
- 82.Klein U., Tu Y., Stolovitzky G.A., Mattioli M., Cattoretti G., Husson H. Gene expression profiling of B cell chronic lymphocytic leukemia reveals a homogeneous phenotype related to memory B cells. J. Exp. Med. 2001;194(11):1625–1638. doi: 10.1084/jem.194.11.1625. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Rosenwald A., Alizadeh A.A., Widhopf G., Simon R., Davis R.E., Yu X. Relation of gene expression phenotype to immunoglobulin mutation genotype in B cell chronic lymphocytic leukemia. J. Exp. Med. 2001;194(11):1639–1647. doi: 10.1084/jem.194.11.1639. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Yu J., Chen L., Cui B., Widhopf G.F., 2nd, Shen Z., Wu R. Wnt5a induces ROR1/ROR2 heterooligomerization to enhance leukemia chemotaxis and proliferation. J. Clin. Invest. 2016;126(2):585–598. doi: 10.1172/JCI83535. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Barna G., Mihalik R., Timar B., Tombol J., Csende Z., Sebestyen A. ROR1 expression is not a unique marker of CLL. Hematol. Oncol. 2011;29(1):17–21. doi: 10.1002/hon.948. [DOI] [PubMed] [Google Scholar]
- 86.Yu J., Chen Y., Chen L., Zhang L., Rassenti L.Z., Widhopf G.F., 2nd Cirmtuzumab inhibits ibrutinib-resistant, Wnt5a-induced Rac1 activation and proliferation in mantle cell lymphoma. Oncotarget. 2018;9(37):24731–24736. doi: 10.18632/oncotarget.25340. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Yu J., Chen L., Cui B., Wu C., Choi M.Y., Chen Y. Cirmtuzumab inhibits Wnt5a-induced Rac1 activation in chronic lymphocytic leukemia treated with ibrutinib. Leukemia. 2017;31(6):1333–1339. doi: 10.1038/leu.2016.368. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Raponi S., De Propris M.S., Intoppa S., Milani M.L., Vitale A., Elia L. Flow cytometric study of potential target antigens (CD19, CD20, CD22, CD33) for antibody-based immunotherapy in acute lymphoblastic leukemia: analysis of 552 cases. Leuk. Lymphoma. 2011;52(6):1098–1107. doi: 10.3109/10428194.2011.559668. [DOI] [PubMed] [Google Scholar]
- 89.Nagorsen D., Kufer P., Baeuerle P.A., Bargou R. Blinatumomab: a historical perspective. Pharmacol. Ther. 2012;136(3):334–342. doi: 10.1016/j.pharmthera.2012.07.013. [DOI] [PubMed] [Google Scholar]
- 90.Gruen M., Bommert K., Bargou R.C. T-cell-mediated lysis of B cells induced by a CD19xCD3 bispecific single-chain antibody is perforin dependent and death receptor independent. Cancer Immunol. Immunother. 2004;53(7):625–632. doi: 10.1007/s00262-003-0496-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91.Molhoj M., Crommer S., Brischwein K., Rau D., Sriskandarajah M., Hoffmann P. CD19-/CD3-bispecific antibody of the BiTE class is far superior to tandem diabody with respect to redirected tumor cell lysis. Mol. Immunol. 2007;44(8):1935–1943. doi: 10.1016/j.molimm.2006.09.032. [DOI] [PubMed] [Google Scholar]
- 92.Brandl C., Haas C., d'Argouges S., Fisch T., Kufer P., Brischwein K. The effect of dexamethasone on polyclonal T cell activation and redirected target cell lysis as induced by a CD19/CD3-bispecific single-chain antibody construct. Cancer Immunol. Immunother. 2007;56(10):1551–1563. doi: 10.1007/s00262-007-0298-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Klinger M., Brandl C., Zugmaier G., Hijazi Y., Bargou R.C., Topp M.S. Immunopharmacologic response of patients with B-lineage acute lymphoblastic leukemia to continuous infusion of T cell-engaging CD19/CD3-bispecific BiTE antibody blinatumomab. Blood. 2012;119(26):6226–6233. doi: 10.1182/blood-2012-01-400515. [DOI] [PubMed] [Google Scholar]
- 94.Bargou R., Leo E., Zugmaier G., Klinger M., Goebeler M., Knop S. Tumor regression in cancer patients by very low doses of a T cell-engaging antibody. Science. 2008;321(5891):974–977. doi: 10.1126/science.1158545. [DOI] [PubMed] [Google Scholar]
- 95.Goebeler M.E., Knop S., Viardot A., Kufer P., Topp M.S., Einsele H. Bispecific T-cell engager (BiTE) antibody construct blinatumomab for the treatment of patients with relapsed/refractory non-hodgkin lymphoma: final results from a phase I study. J. Clin. Oncol. 2016;34(10):1104–1111. doi: 10.1200/JCO.2014.59.1586. [DOI] [PubMed] [Google Scholar]
- 96.Gupta P., Goldenberg D.M., Rossi E.A., Cardillo T.M., Byrd J.C., Muthusamy N. Dual-targeting immunotherapy of lymphoma: potent cytotoxicity of anti-CD20/CD74 bispecific antibodies in mantle cell and other lymphomas. Blood. 2012;119(16):3767–3778. doi: 10.1182/blood-2011-09-381988. [DOI] [PubMed] [Google Scholar]
- 97.Davis T.A., Czerwinski D.K., Levy R. Therapy of B-cell lymphoma with anti-CD20 antibodies can result in the loss of CD20 antigen expression. Clin. Cancer Res. 1999;5(3):611–615. [PubMed] [Google Scholar]
- 98.Haidar J.H., Shamseddine A., Salem Z., Mrad Y.A., Nasr M.R., Zaatari G. Loss of CD20 expression in relapsed lymphomas after rituximab therapy. Eur. J. Haematol. 2003;70(5):330–332. doi: 10.1034/j.1600-0609.2003.00007.x. [DOI] [PubMed] [Google Scholar]
- 99.Alvaro-Naranjo T., Jaen-Martinez J., Guma-Padro J., Bosch-Princep R., Salvado-Usach M.T. CD20-negative DLBCL transformation after rituximab treatment in follicular lymphoma: a new case report and review of the literature. Ann. Hematol. 2003;82(9):585–588. doi: 10.1007/s00277-003-0694-1. [DOI] [PubMed] [Google Scholar]
- 100.Hiraga J., Tomita A., Sugimoto T., Shimada K., Ito M., Nakamura S. Down-regulation of CD20 expression in B-cell lymphoma cells after treatment with rituximab-containing combination chemotherapies: its prevalence and clinical significance. Blood. 2009;113(20):4885–4893. doi: 10.1182/blood-2008-08-175208. [DOI] [PubMed] [Google Scholar]
- 101.Duman B.B., Sahin B., Ergin M., Guvenc B. Loss of CD20 antigen expression after rituximab therapy of CD20 positive B cell lymphoma (diffuse large B cell extranodal marginal zone lymphoma combination): a case report and review of the literature. Med. Oncol. 2012;29(2):1223–1226. doi: 10.1007/s12032-011-9955-3. [DOI] [PubMed] [Google Scholar]
- 102.Beum P.V., Kennedy A.D., Williams M.E., Lindorfer M.A., Taylor R.P. The shaving reaction: rituximab/CD20 complexes are removed from mantle cell lymphoma and chronic lymphocytic leukemia cells by THP-1 monocytes. J. Immunol. 2006;176(4):2600–2609. doi: 10.4049/jimmunol.176.4.2600. [DOI] [PubMed] [Google Scholar]
- 103.Beers S.A., French R.R., Chan H.T., Lim S.H., Jarrett T.C., Vidal R.M. Antigenic modulation limits the efficacy of anti-CD20 antibodies: implications for antibody selection. Blood. 2010;115(25):5191–5201. doi: 10.1182/blood-2010-01-263533. [DOI] [PubMed] [Google Scholar]
- 104.Jazirehi A.R., Vega M.I., Bonavida B. Development of rituximab-resistant lymphoma clones with altered cell signaling and cross-resistance to chemotherapy. Cancer Res. 2007;67(3):1270–1281. doi: 10.1158/0008-5472.CAN-06-2184. [DOI] [PubMed] [Google Scholar]
- 105.Cragg M.S., Glennie M.J. Antibody specificity controls in vivo effector mechanisms of anti-CD20 reagents. Blood. 2004;103(7):2738–2743. doi: 10.1182/blood-2003-06-2031. [DOI] [PubMed] [Google Scholar]
- 106.Janas E., Priest R., Wilde J.I., White J.H., Malhotra R. Rituxan (anti-CD20 antibody)-induced translocation of CD20 into lipid rafts is crucial for calcium influx and apoptosis. Clin. Exp. Immunol. 2005;139(3):439–446. doi: 10.1111/j.1365-2249.2005.02720.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107.Bonavida B. Rituximab-induced inhibition of antiapoptotic cell survival pathways: implications in chemo/immunoresistance, rituximab unresponsiveness, prognostic and novel therapeutic interventions. Oncogene. 2007;26(25):3629–3636. doi: 10.1038/sj.onc.1210365. [DOI] [PubMed] [Google Scholar]
- 108.Cartron G., Dacheux L., Salles G., Solal-Celigny P., Bardos P., Colombat P. Therapeutic activity of humanized anti-CD20 monoclonal antibody and polymorphism in IgG Fc receptor FcgammaRIIIa gene. Blood. 2002;99(3):754–758. doi: 10.1182/blood.v99.3.754. [DOI] [PubMed] [Google Scholar]
- 109.Kim D.H., Jung H.D., Kim J.G., Lee J.J., Yang D.H., Park Y.H. FCGR3A gene polymorphisms may correlate with response to frontline R-CHOP therapy for diffuse large B-cell lymphoma. Blood. 2006;108(8):2720–2725. doi: 10.1182/blood-2006-01-009480. [DOI] [PubMed] [Google Scholar]
- 110.Ahlgrimm M., Pfreundschuh M., Kreuz M., Regitz E., Preuss K.D., Bittenbring J. The impact of Fc-gamma receptor polymorphisms in elderly patients with diffuse large B-cell lymphoma treated with CHOP with or without rituximab. Blood. 2011;118(17):4657–4662. doi: 10.1182/blood-2011-04-346411. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111.Galimberti S., Palumbo G.A., Caracciolo F., Benedetti E., Pelosini M., Brizzi S. The efficacy of rituximab plus Hyper-CVAD regimen in mantle cell lymphoma is independent of FCgammaRIIIa and FCgammaRIIa polymorphisms. J. Chemother. 2007;19(3):315–321. doi: 10.1179/joc.2007.19.3.315. [DOI] [PubMed] [Google Scholar]
- 112.Cheah C.Y., Seymour J.F., Wang M.L. Mantle cell lymphoma. J. Clin. Oncol. 2016;34(11):1256–1269. doi: 10.1200/JCO.2015.63.5904. [DOI] [PubMed] [Google Scholar]

