Abstract
With the introduction of anti‐CD19 chimeric antigen receptor (CAR) T‐cell (CAR T) therapies, bispecific CD3/CD20 antibodies and anti‐CD19 antibodies, immunotherapy continues to transform the treatment of diffuse large B‐cell lymphoma (DLBCL). A number of novel immunotherapeutic strategies are under investigation to build upon current clinical benefit and offer further options to those patients who cannot tolerate conventional intensive therapies due to their age and/or state of health. Alongside immunotherapies that leverage the adaptive immune response, natural killer (NK) cell and myeloid cell‐engaging therapies can utilize the innate immune system. Monoclonal antibodies engineered for greater recognition by the patient's immune system can enhance antitumor cytotoxic mechanisms mediated by NK cells and macrophages. In addition, CAR technology is extending into NK cells and macrophages and investigational immune checkpoint inhibitors targeting macrophage/myeloid cell checkpoints via the CD47/SIRPα axis are in development. Regimens that engage both innate and adaptive immune responses may help to overcome resistance to current immunotherapies. Furthermore, combinations of immunotherapy and oncogenic pathway inhibitors to reprogram the immunosuppressive tumor microenvironment of DLBCL may also potentiate antitumor responses. As immunotherapy treatment options continue to expand, both in the first‐line setting and further lines of therapy, understanding how to harness these immunotherapies and the potential for combination approaches will be important for the development of future DLBCL treatment approaches.
Keywords: diffuse large B‐cell lymphoma, DLBCL

1. BACKGROUND
First‐line R‐CHOP (rituximab plus cyclophosphamide, doxorubicin, vincristine, and prednisone) and similar regimens are curative in 60%–70% of patients with diffuse large B‐cell lymphoma (DLBCL), with 30%–40% of patients experiencing relapse or a primary refractory disease course. 1 However, some patients are unable to tolerate the R‐CHOP regimen due to older age, being unfit/frail, and/or having comorbidities. 2 , 3 Furthermore, some patients are at high risk of relapse/refractory (R/R) disease following first‐line R‐CHOP due to higher International Prognostic Index (IPI) score 4 or high‐grade lymphoma classification (MYC plus BCL2 and/or BCL6 rearrangements, also known as double or triple hit lymphomas). 5 Effective treatment options for these patients are currently lacking.
A number of treatment strategies have been evaluated to improve the efficacy of first‐line therapy, or provide novel options for patients who are frail/elderly. Intensified chemotherapy approaches have not improved upon the R‐CHOP first‐line standard of care and have been associated with increased toxicity. 6 , 7 , 8 In older and/or unfit patients with DLBCL, approaches to improve tolerability of anthracycline‐based chemotherapy may compromise efficacy. Rituximab with dose‐reduced CHOP (R‐miniCHOP) 9 and ofatumumab (anti‐CD20)‐miniCHOP 10 in patients older than 80 years with DLBCL and who have a good performance status improved tolerability and provided an effective treatment option, albeit with response and survival rates lower than in younger patients with DLBCL. The addition of targeted therapies such as Bruton's tyrosine kinase inhibitors (BTKis) and BCL2 inhibitors to first‐line R‐CHOP have similarly met with limited success, failing to show significant efficacy improvements and are associated with increased toxicity. 11 , 12 , 13 , 14 The use of targeted chemotherapy in the form of antibody–drug conjugates (ADCs) has proven a more successful strategy. The Phase III POLARIX study evaluating a modified regimen of R‐CHOP (pola‐R‐CHP), in which vincristine was replaced with polatuzumab vedotin (an ADC targeting CD79b) in patients with previously untreated intermediate‐risk or high‐risk DLBCL, showed a statistically significant slightly lower risk of disease progression, relapse or death compared to R‐CHOP. 15
Immunomodulation with lenalidomide, which has been shown to block tumor cell proliferation and angiogenesis, and stimulate T‐ and NK cell‐mediated cytotoxicity, has also been evaluated as a strategy to improve current DLBCL treatment options, but has met with mixed results. Lenalidomide in combination with R‐CHOP (also known as R2‐CHOP) had numerically greater progression‐free survival (PFS) and overall survival (OS) compared to R‐CHOP; however, the study was not powered adequately for definitive comparisons. 16 The Phase III ROBUST study, in which a lower dose of lenalidomide was used, did not meet the primary endpoint of PFS but did report a nonsignificant positive trend in 2‐year PFS rate in a subgroup analysis of patients with high‐risk disease (IPI scores ≥3 and advanced disease stages). 17 In elderly and unfit/frail patients, a chemotherapy‐free option of lenalidomide plus rituximab has been assessed as an alternative to standard first‐line anthracycline‐containing regimens. Although the Phase II ReRI study did not meet its primary efficacy endpoint, the observed clinical activity warrants further exploration of this combination. 18
Immunotherapy has revolutionized the treatment of R/R DLBCL through the introduction of anti‐CD19 antibodies, anti‐CD19 CAR T‐cell (CAR T) therapies, and bispecific CD3/CD20 antibodies (Figure 1). While these approaches have shown significant clinical benefit in R/R DLBCL and hold promise for patients with treatment‐naïve and high‐risk DLBCL, there are still challenges to overcome. Looking to the future of DLBCL management, utilizing novel immunotherapies that harness the immune system will be critical to addressing the current unmet need. The objective of this review is to outline the most relevant immune pathways and promising agents that target them, and explore the potential synergies offered by combination approaches (Figure 2).
FIGURE 1.

A timeline of the era of immunotherapy for DLBCL. CAR T‐cell, chimeric antigen receptor T‐cell; CELMoD, cereblon E3 ligase modulator; CHOP, cyclophosphamide, doxorubicin, vincristine, and prednisone; DLBCL, diffuse large B‐cell lymphoma; FDA, US Food and Drug Administration; NK, natural killer.
FIGURE 2.

Current and future immunotherapeutic treatments for DLBCL. ADCC, antibody‐dependent cellular cytotoxicity; ADCP, antibody‐dependent cellular phagocytosis; BTK, Bruton's tyrosine kinase; CAR T‐cell, chimeric antigen receptor T‐cell; DLBCL, diffuse large B‐cell lymphoma; ITIM, immunoreceptor tyrosine‐based inhibitory motif; NK, natural killer; TME, tumor microenvironment.
2. IMMUNOTHERAPIES UTILIZE A RANGE OF APPROACHES TO SUPPORT THE IMMUNE SYSTEM TO ELIMINATE CANCER CELLS
Both myeloid cells of the innate immune system and lymphocytes of the adaptive immune response have a critical role in immune surveillance and control of B‐cell lymphomas. T cells primarily act either as cytotoxic CD8+ T cells attacking cells or as CD4 helper T cells (CD4+ T cells) supporting B‐cell antigen production. T cells are classically activated by an interaction between the T‐cell receptor (TCR) and a major histocompatibility complex (MHC)‐bound antigen on antigen‐presenting cells, 19 resulting in a complex cascade of activation pathways. The key antitumor effector mechanism mediated by CD8+ T cells that is exploited by several immunotherapeutic strategies is perforin/granzyme‐mediated cell death. Mobilization and activation of CD4+ T cells is also crucial to mounting an effective antitumor response in DLBCL, as patients with DLBCL and >20% infiltrating CD4+ T cells have better relapse‐free survival and OS. 20 , 21 However, 50% of DLBCL cases lack cell surface expression of MHC Class I 22 with loss of MHC Class II expression also reported, 23 both of which compromise effective tumor immunosurveillance.
NK cells of the innate immune system target cancer cells through the detection of MHC Class I molecule downregulation or stress markers. 24 Activation of their cytotoxicity, cytokine production, and proliferation is mediated via a balance of signals from activating and inhibitory receptors, such as FcγRIII (CD16), NKG2D, killer cell immunoglobulin‐like receptors and immune checkpoint molecules. 24 Several immunotherapeutic strategies exploit the potent cytotoxic function of NK cells in an MHC‐independent manner through FcγRIII (CD16) engagement and subsequent antibody‐dependent cellular cytotoxicity (ADCC). Myeloid cells, comprising macrophages, dendritic cells, monocytes, and granulocytes, function as key antigen‐presenting cells and play a pivotal part in tumor clearance through macrophage‐mediated antibody‐dependent cellular phagocytosis (ADCP) following chemotherapy and immunotherapy. Myeloid cells represent a significant proportion of the tumor immune microenvironment in DLBCL 25 and can create an immune suppressive or stimulatory environment that positively or negatively influences cancer progression and metastasis. 26 Modulating myeloid cell functions to create a stimulatory, antitumor environment may augment the efficacy of current and novel treatment options and provides a new avenue for the development of immunotherapeutic strategies.
Several novel therapies based on T‐cell, NK‐cell or myeloid cell‐engaging or ‐derived therapies are under development. Thus far, these emerging immunotherapies are primarily under investigation in the R/R DLBCL disease setting, with the hope of a widening scope to also improve first‐line options.
2.1. T cell‐engaging or ‐derived immunotherapies
2.1.1. CAR T‐cell therapies
CAR T‐cell therapies are derived from blood cells modified in vitro using viral transduction to express a CAR on the cell surface, which can target a tumor antigen. The most common current version of CAR is an anti‐CD19 single‐chain variable fragment linked to an intracellular signaling domain (usually CD3ζ), resulting in MHC‐independent T‐cell activation upon binding CD19 on the surface of malignant B cells and triggering cytotoxicity. 27 In DLBCL, a number of autologous CAR T‐cell therapies have been approved in the third‐line R/R disease setting: axicabtagene ciloleucel (axi‐cel; ZUMA‐1 28 , 29 ), tisagenlecleucel (tisa‐cel, JULIET 30 ), and lisocabtagene maraleucel (liso‐cel, TRANSCEND‐NHL‐001 31 ). These third‐line CAR T‐cell therapies have shown robust and long‐term responses in DLBCL, and demonstrated similar efficacy in several real‐world studies. 32 More recently, the use of second‐line CAR T‐cell therapy has been approved following data from the ZUMA‐7 and TRANSFORM/PILOT studies. 33 , 34 , 35 The high response rates and the durability of the responses have led to the initiation of a number of studies in treatment‐naïve patients with high‐risk LBCL (ZUMA‐12 36 ; ZUMA‐23 37 ).
Practical considerations with CAR T‐cell therapy include treatment center resources and potential immune‐mediated side effects. 32 The requirement for autologous T cells for the manufacture of the treatment product may also limit patient eligibility as many patients are heavily pretreated and may have unfit T cells. 38 Due to the CAR T‐cell manufacturing time, bridging therapy may be required; however, the use of bridging therapy has shown to be associated with worsened outcomes, likely due in part to the reasons why bridging is required such as aggressive disease. 39 CAR T‐cell therapies are also associated with toxicities such as cytokine‐release syndrome (CRS) and neurotoxicities. 40 , 41
Continued development in the CAR T‐cell therapy space is focused on improvement of treatment efficacy. Due to the immunosuppressive tumor microenvironment (TME) in patients with solid tumors, a barrier is created, limiting the long‐term efficacy of CAR T‐cell therapy in these patients, resulting in further cancer progression. 42 , 43 , 44 One method under consideration to address this issue is the creation of armored CAR T cells, which are engineered to express proteins alongside the CAR in an effort to reduce immunosuppression and improve antitumor efficacy. 42 , 43 , 44 Armored CAR T‐cell therapy includes TRUCK (T cells Redirected toward Universal Cytokine Killing) CARs, cytokine modulating CARs and antibody‐secreting CARs. Another emerging approach, potentially with more immediate relevance to patients with DLBCL, uses CAR T cells directed at multiple B‐cell antigens, rather than just CD19, such as CD20 and CD22, which has the potential to improve response by preventing antigen escape. 45 Investigational candidate IMPT‐314, a CD19/20 bispecific CAR T‐cell therapy, is in Phase I/II development in patients with R/R B‐cell non‐Hodgkin lymphoma (R/R B‐NHL; NCT05826535). 46
“Off‐the‐shelf” allogeneic CAR T cells have several potential advantages over autologous CAR T cells, including shorter manufacturing time, greater manufacturing scalability, and avoidance of poor T‐cell “fitness” from heavily pretreated patients. Despite their potential advantages, allogeneic products may also present specific issues such as short persistence due to rapid elimination by the immune system, along with other risks such as graft‐versus‐host disease (GvHD) and graft rejection, which can be minimized with genomic editing during allogeneic CAR T‐cell manufacture. 47 A number of allogeneic CAR T cells are under clinical investigation. 48 The most common adverse events associated with allogeneic CAR T cells are mild‐to‐moderate cytopenias, CRS and neurotoxicity, with negligible incidences of GvHD reported. 49
2.1.2. Bispecific and trispecific antibodies
Bispecific antibodies are a promising T‐cell‐mediated approach which use the specificity of monoclonal antibodies to engage the patient's own T cells to kill malignant B cells. Engineered by combining two different monoclonal antigen‐specific binding regions from different antibodies into one molecule, they simultaneously target both a tumor cell and a T‐cell epitope. 50
In DLBCL, the majority of bispecific antibodies currently in advanced clinical development target CD20 on B cells and CD3 of the TCR complex, bypassing MHC/TCR binding and resulting in T‐cell engagement. 51 CD3–CD20 bispecific antibodies epcoritamab and glofitamab recently received regulatory approval in the United States, and Canada and Europe, respectively, for R/R DLBCL, with mosunetuzumab and odronextamab in advanced clinical development in patients with R/R disease. Bispecific antibodies in combination with CHOP+/−R are also under evaluation as first‐line therapy. 50 , 51 , 52 TNB‐486 is a novel CD3–CD19 bispecific antibody in Phase I development in R/R DLBCL. 53
As with CAR T‐cell therapy, CRS has been shown to be associated with bispecific antibodies, but generally to a lower incidence and severity than with CAR T. 51 To further mitigate potential for CRS, “step‐up dosing,” subcutaneous formulations, or the use of a cytoreductive anti‐CD20 antibody prior to administration of the bispecific therapy are being evaluated. 54
Taking the concept of multitargeting introduced with bispecific antibodies a step further, trispecific antibodies are at an early stage of investigation, with PIT565 demonstrating simultaneous engagement of CD19 on tumor cells, CD3 and CD2 on T cells leading to redirected T‐cell cytotoxicity toward CD19+ malignant B cells. Preclinical data have suggested the potential for deeper and more durable responses compared to CD3 bispecifics and a Phase I investigation is underway in patients with R/R B‐NHL and R/R B‐cell acute lymphoblastic leukemia. 55 A further trispecific antibody, JNJ‐8543 (JNJ‐80948543), is also being investigated in a Phase I trial across a range of indications, including DLBCL. 56
2.1.3. Beyond T‐cell‐engaging or ‐derived immunotherapies
While long‐term efficacy data for bispecific antibodies are just beginning to emerge, 57 recently reported long‐term efficacy data from studies of CAR T‐cell therapies show that ~60%–70% of patients experience a relapse within 2–3 years of treatment initiation. 29 , 30 , 58 These results suggest that additional therapeutic options are needed. Combination therapy utilizing the innate immune system via NK cell and myeloid cell‐engaging or ‐derived therapies may improve upon current immunotherapies.
2.2. NK cell and myeloid cell‐engaging or ‐derived therapies
2.2.1. Monoclonal antibodies
Rituximab, the CD20‐targeting monoclonal antibody introduced the significant role for monoclonal antibody immunotherapy in B‐cell NHL. Monoclonal antibodies bind to their epitope whereby the fragment crystallizable (Fc) domain is then recognized by the patient's immune system, triggering antitumor cytotoxic mechanisms (ADCC, ADCP, and complement‐dependent cytotoxicity) largely mediated by NK and myeloid cells.
In treatment‐naïve patients with DLBCL, ~15%–20% have low CD20 expressing tumors. 59 , 60 CD19, however, is ubiquitously expressed throughout the B‐cell maturation process and is broadly conserved across many B‐cell malignancies. 59 , 60 , 61 , 62 Tafasitamab is an anti‐CD19, humanized, monoclonal antibody with an engineered Fc region for increased affinity for FcγRIII, leading to increased ADCC and ADCP by NK cells and macrophages. 63 , 64 ADCC and ADCP are further enhanced when tafasitamab is used in combination with lenalidomide, which induces FcγRIII expression on NK cells, allowing for a greater degree of activation following tafasitamab binding. 65 Tafasitamab plus lenalidomide has shown an overall response rate of almost 60% and a duration of response of 43.9 months in patients with R/R DLBCL in the L‐MIND study, with a 5‐year analysis demonstrating long‐term durable responses (median duration of response not reached). 66 , 67
2.2.2. NK cells
Therapeutic monoclonal antibody approaches in DLBCL rely on the engagement of NK cells via FcγRIII (CD16) to facilitate ADCC of targeted cells. Due to their intrinsic cytotoxic capacity and low potential for autoreactivity inherent with T‐cell approaches, NK cells themselves are attractive options for cellular immunotherapy.
NK cells that have been expanded and activated, although otherwise unmodified, do not require antigen presentation (unlike T‐cell‐based therapies) when used as an adoptive cell therapy. 68 Autologous and haploidentical NK cells have been investigated alone and in combination with various other therapies in several hematological cancers. From a safety perspective, allogeneic NK cells benefit from limited reported potential toxicity issues compared to allogeneic CAR T cells, including GVHD.
CAR NK cells could offer several advantages over CAR T cells, such as rapid generation from multiple allogeneic sources offering an “off‐the‐shelf” option for patients with heavily pretreated or rapidly progressing disease, reduced risk of alloreactivity inherent to NK cells and a superior safety profile in terms of little to no reported incidence of CRS or neurotoxicity. 69 , 70 , 71 A recent Phase I/II trial confirmed the preliminary efficacy and tolerability of CAR NK cells derived from cord blood in heavily pretreated patients with CD19‐positive lymphoid cancers including two patients with refractory DLBCL. 69 Clinical trials are ongoing to further evaluate the safety and efficacy of this treatment approach. Interestingly, a novel approach utilizing cytokine‐induced cord blood‐derived NK cells precomplexed with the CD30/CD16 bispecific antibody, AFM13, demonstrated CAR NK cell‐like features in preclinical studies and clinical activity in a proof‐of‐concept study in patients with R/R CD30+ Hodgkin's or non‐Hodgkin's lymphoma. 72
2.2.3. CD47/SIRPα axis
The CD47/SIRPα axis is an important checkpoint, mediating antitumor responses by the innate immune system. CD47 is ubiquitously expressed and binds to SIRPα, an inhibitory receptor expressed on the surface of macrophages, to inhibit macrophage recognition and subsequent phagocytosis. 73 , 74 In DLBCL, overexpression of CD47 was associated with worse clinical outcomes in patients with DLBCL treated with CHOP±R. 75 Exploitation of the CD47/SIRPα axis by blocking CD47‐SIRPα signaling represents a promising therapeutic strategy with several Phase I/II studies underway evaluating anti‐CD47 monoclonal antibodies (magrolimab [Hu5F9‐G4]; ligufalimab [AK117]; lemzoparlimab [TJC4]; AO‐176) or anti‐SIRPα Fc‐fusion proteins (evorpacept [ALX148]; TTI‐621; TTI‐622; IMM01).
2.2.4. CAR macrophages
CAR technology has also extended to macrophages, wherein preclinical models suggest CAR macrophages have potential advantages over traditional CAR T cells in terms of efficiently redirecting macrophages, guiding antigen‐dependent phagocytosis, and potentiating antitumor T‐cell activity. 76 CAR macrophages are currently only in clinical development in solid tumors.
3. POTENTIAL FOR NOVEL COMBINATION THERAPIES
3.1. B‐cell surface marker combinations
Targeting two or more B‐cell surface markers simultaneously may limit resistance via antigen loss. 59 , 60 , 77 Targeting CD19 and CD20 simultaneously with tafasitamab and rituximab, supplemented by lenalidomide to enhance ADCC activity, in combination with CHOP has demonstrated promising clinical activity as first‐line therapy in the Phase Ib First‐MIND study, with no new adverse safety signals to what would be expected. 78 First‐line tafasitamab and lenalidomide plus R‐CHOP is currently under further investigation in the ongoing, fully enrolled Phase III frontMIND study (NCT04824092). Glofitamab in combination with R‐CHOP or pola‐R‐CHP is being evaluated in treatment‐naïve patients with DLBCL (NCT04914741). 79 In the R/R DLBCL setting, polatuzumab vedotin plus rituximab‐chemotherapy combinations have shown to be effective treatment combinations. 80 The combination of polatuzumab vedotin and mosunetuzumab is being studied in the Phase III SUNMO trial in patients with R/R DLBCL. 81
Enhanced NK cell‐mediated ADCC evident in preclinical data 65 is utilized in L‐MIND with the combination of tafasitamab and lenalidomide 66 illustrating the potential for immunomodulatory drugs to boost current monoclonal antibody therapy activity. TAK‐981, a small molecule inhibitor of the small ubiquitin‐like modifier (SUMO), brings in another synergistic approach, as a potent and selective inhibitor of SUMOylation. This reversible posttranslational modification results in an inflammatory response mediated by type 1 interferons, increased NK cell activation and M1 macrophage polarization. 82 Combinations of tafasitamab and rituximab with TAK‐981 were found to markedly enhance ADCC and ADCP and augment antitumor activity compared to the respective monotherapies. 82 Immunomodulation of the adaptive immune response is being evaluated with the combination of epcoritamab with lenalidomide as first‐line therapy for DLBCL. 83
With the availability of an array of immunotherapy modalities targeting cell surface molecules, a broad range of combination approaches are now being assessed with the objective of developing chemotherapy‐free approaches. The Phase II Smart Stop study (NCT04978584) is assessing the combination of tafasitamab, rituximab, lenalidomide, and the BTKi acalabrutinib as an immunotherapy combination alone or with CHOP in patients with treatment‐naïve nongerminal center (non‐GCB) DLBCL. 84 This trial aims to build on the proof‐of‐concept Smart Start trial (NCT02636322), which showed that the combination of rituximab, lenalidomide, and ibrutinib followed by sequential addition of chemotherapy led to high overall response rates and durable responses compared to historical outcomes, 85 highlighting the feasibility of such an approach. Other chemotherapy‐free combination approaches with monoclonal antibodies, ADCs, and bispecific antibodies are being explored. 86 Mosunetuzumab is being investigated as consolidation therapy and as monotherapy or in combination with polatuzumab vedotin in elderly/unfit patients with previously untreated DLBCL (NCT03677154).
A phase II study of mosunetuzumab, polatuzumab, tafasitamab, and lenalidomide in patients with relapsed B‐cell NHL is ongoing (NCT05615636). 87 Interestingly, the sequence of the immunotherapy combinations may be a consideration. Preclinical data suggest the sequential use of tafasitamab followed by CD19‐targeted CAR T‐cell therapy in xenograft models ameliorated side effects of T‐cell overactivation and promoted antitumor activity of CAR T‐cell therapy. 88 , 89 Loncastuximab tesirine, an ADC targeting CD19, was being evaluated in combination with rituximab in a Phase II trial in previously untreated unfit/frail patients with DLBCL; however, the study was terminated due to adverse respiratory safety signals (LOTIS‐9; NCT05144009). 86 , 90 , 91 Other potential treatment options include cereblon E3 ligase modulators (CELMoDs) that are being investigated in combination with immunotherapeutics. A phase I/II study investigating the CELMoD golcadomide alone and in combination with rituximab in patients with R/R DLBCL or R/R follicular lymphoma (FL) is currently underway (NCT03930953). 92 Preliminary results have shown that golcadomide plus rituximab has a similar safety profile to golcadomide alone and has shown promising efficacy outcomes. Another CELMoD, avadomide, has completed a phase I trial in combination with rituximab in patients with R/R DLBCL or R/R FL, with positive safety outcomes and preliminary antitumor activity reported (NCT03283202). 93 Other CELMoD candidates are under investigation, and may eventually be used in combination with immunotherapies; however, these agents are outside of the scope of the current review.
3.2. Immune checkpoint inhibition
A recent systematic review and meta‐analysis reported limited clinical benefit with immune checkpoint inhibitors (ICIs) targeting programmed cell death protein 1 (PD‐1) and its ligand (PD‐L1) or cytotoxic T‐lymphocyte‐associated protein 4 (CTLA‐4) as monotherapy in R/R DLBCL. 94 Inhibition of alternative immune checkpoint molecules is being explored to enhance immunostimulatory and antitumor activity in DLBCL, most notably T‐cell immunoreceptor with Ig and ITIM domains (TIGIT), lymphocyte‐activated gene‐3 (LAG‐3), and T‐cell immunoglobulin and mucin‐containing protein‐3 (TIM‐3), all of which mediate suppression of T‐cell activation. Several early‐phase studies are underway to evaluate the blockade of these immune checkpoint molecules in DLBCL. 95 , 96 , 97 , 98 , 99 , 100
While most ICI combinations focus on T‐cell effector function, inhibitors of the macrophage/myeloid cell checkpoint via the CD47/SIRPα axis are also being studied as a combination partner to further enhance the efficacy of DLBCL immunotherapy. Magrolimab in combination with rituximab has shown promising activity with rapid and durable responses in a Phase Ib/II study in patients with R/R DLBCL. 101 , 102 Preclinical data show combining tafasitamab with an anti‐CD47 monoclonal antibody increases the ADCP activity of primary macrophages, resulting in enhanced antitumor activity compared to tafasitamab or anti‐CD47 monotherapies alone. 103 , 104 To further maximize the immunotherapy combination of tafasitamab plus lenalidomide, the addition of TTI‐622 is being studied in a Phase Ib/II study in patients with R/R DLBCL (NCT05626322). A similar study is ongoing for ALX148, rituximab, and lenalidomide for the treatment of indolent and aggressive B‐NHL (NCT05025800). The potential for a combined antagonism of CD47/SIRPα and PD‐1/PD‐L1 pathways is being evaluated in a Phase II study of TTI‐622 and TTI‐621 in combination with pembrolizumab for the treatment of R/R DLBCL (NCT05507541).
3.3. Tumor microenvironment reprogramming/remodeling
Tumor antigen heterogeneity, limited immune cell trafficking and infiltration, T‐cell exhaustion, MHC downregulation, and anti‐inflammatory cytokines are all key features of the “noninflamed” or immunosuppressive DLBCL tumor microenvironment (TME). 20 , 105 , 106 , 107 Reprogramming or remodeling the TME across a variety of immunological parameters using myeloid and T‐cell‐based approaches may potentially enhance long‐term responses.
3.3.1. Enhancing immune cell infiltration
DLBCL is characterized by low immune cell infiltration, especially in the GCB subtype. 20 , 107 , 108 The presence of tumor‐infiltrating lymphocytes is indicative of immune‐inflamed tumors and an active antitumor immune response. 107 Tumor infiltration of CD4 T+ cells may shift the balance of suppressive and pro‐inflammatory cytokines toward a TME more conducive to infiltration of cytotoxic T cells or CAR T cells. NK cell and myeloid cell‐engaging or ‐derived therapies have the potential to produce pro‐inflammatory cytokines and chemokines to recruit other immune cells, such as CAR T cells and macrophages. 76 , 109 , 110
3.3.2. Switching to a pro‐inflammatory cytokine tumor microenvironment
A challenge for maintaining immunotherapy responses in DLBCL is the presence of anti‐inflammatory cytokines, such as TGF‐β, interleukin‐10 (IL‐10), and vascular endothelial growth factor (VEGF), principally produced by tumor‐associated macrophages (TAMs), myeloid‐derived suppressor cells, and cancer‐associated fibroblasts. 105 , 107 Modulating the cytokine environment through allogeneic or CAR NK cells, and potentially CAR macrophages, may enhance immunotherapy responses by promoting an immune‐inflamed TME to maintain immune cell activation. 76 , 109 , 110
3.3.3. Addressing T‐cell exhaustion
In addition to the immunosuppressive TME, chronic or persistent T‐cell exposure to tumor antigens is associated with marked changes in T‐activation and differentiation, leading to T‐cell “dysfunction” or “exhaustion”, defined by poor effector function, sustained expression of inhibitory receptors, and a transcriptional state different to that of functional effector or memory T cells. 111 , 112 In DLBCL, T‐cell exhaustion may be a mechanism for DLBCL progression after treatment with indirect T cell‐engaging immunotherapy, bispecific antibodies, or CAR T‐cell therapies, particularly if multiple T‐cell approaches are used. 111 Expression of inhibitory checkpoint molecules, such as PD‐1, CTLA‐4, LAG‐3, TIGIT, and TIM‐3 are phenotypically characteristic of exhausted T cells. 38 , 111 , 113 , 114 , 115 Variability in CAR T‐cell efficacy may be linked to heterogeneity in the cellular and molecular features of CAR T‐cell infusion products with the inclusion of T cells with exhausted characteristics, particularly high expression of LAG‐3 and TIM‐3. 38 T cells may be reactivated or activation maintained with simultaneous or optimally sequenced approaches.
3.3.4. Oncogenic signaling pathway inhibition within the TME
Molecular profiling of the TME identifies several genomic and oncological drivers that can be therapeutically targeted alongside immune cell strategies and lead to novel combinations. 116 Targeting the B‐cell receptor and its intracellular signaling pathways presents a logical rationale for combination with immunotherapy. Indeed, data from the Smart Start and Smart Stop studies suggest the inclusion of BTKis with immunotherapy to be a promising approach. Inhibitors of the phosphatidylinositol 3 kinase (PI3K) signaling pathway can promote T‐cell activation and inhibit immunosuppressive immune cell populations. 116 Bcl2 is a characteristic of GCB‐derived, high‐grade B‐cell lymphomas, and studies have already reported for venetoclax in DLBCL. However, as with BTKis and PI3K inhibitors, these studies have been conducted thus far in combination with chemoimmunotherapeutic regimens and are often associated with notable toxicities, probably underlying the biological complexity and requiring greater optimization for effective combination with immunotherapy. 117 Aberrant epigenetic programming affecting the lymphoma TME has been reported, with epigenetic markers enriched in DLBCL, notably the GCB phenotype. 108 , 118 , 119 EZH2‐activating mutations are associated with MHC downregulation in DLBCL. 120 EZH2 inhibition with tazemetostat or tulmimetostat, in addition to targeting other epigenetic mechanisms with histone deacetylase inhibitors, may play a role in modulating the TME by restoring immune signaling. 119 The hypoxic state of the DLBCL TME plays a fundamental role in immune evasion as hypoxia can induce the expression of immunosuppressive factors, such as VEGF, induce the expression of inhibitory immune checkpoints such as PD‐(L)1, LAG‐3, TIM‐3, CTLA‐4, and CD47 121 and modulate pro‐tumorigenic TAMs. 122 Combinations of immunotherapy and therapies targeting hypoxia inducible factor‐1α/VEGF, PI3K, or mitogen‐activated protein kinase signaling pathways may offer novel therapeutic approaches to overcome limitations and maximize antitumor immunity.
4. CONCLUSIONS
Advances in immunotherapy will continue to transform the future of DLBCL therapy. Combination therapy leveraging both the innate and adaptive immune systems simultaneously could overcome known immunotherapy “resistance” mechanisms to current T‐cell‐derived immunotherapies (e.g., loss of antigen, T‐cell exhaustion, immunosuppressive TME). Novel NK cell and myeloid cell‐engaging therapy combinations may further improve clinical benefit, not only in the R/R setting, but also in the first‐line setting where chemotherapy has not compromised the patient's immune system. Furthermore, chemotherapy‐free options offer treatments with more tolerable safety profiles for patients who are elderly or frail/unfit and unable to tolerate conventional intensive chemotherapies.
AUTHOR CONTRIBUTIONS
Johannes Duell: Conceptualization; writing—review and editing. Jason Westin: Conceptualization; writing—review and editing.
FUNDING INFORMATION
This work was supported by MorphoSys AG and Incyte Corporation.
CONFLICT OF INTEREST STATEMENT
Johannes Duell declares: Nothing to disclose. Jason Westin declares: Research funding from: ADC Therapeutics, Allogene, BMS, Genentech, Janssen, Kite/Gilead, MorphoSys/Incyte, Novartis, Nurix; Consulting funding from: AbbVie, ADC Therapeutics, Allogene, BMS, Genentech, Genmab, Janssen, Kite/Gilead, MorphoSys/Incyte, Novartis, Nurix, and Pfizer.
ACKNOWLEDGMENTS
We thank Stuart Langley of Syneos Health, UK and Envision Pharma Group (Fairfield, CT, USA) for editorial, graphical and writing support. Open Access funding enabled and organized by Projekt DEAL.
Duell J, Westin J. The future of immunotherapy for diffuse large B‐cell lymphoma . Int J Cancer. 2025;156(2):251‐261. doi: 10.1002/ijc.35156
MorphoSys AG and Incyte Corporation conducted courtesy reviews for scientific accuracy of this article prior to submission but had no role in the development of the content, selection of journal for submission, or the decision to submit the article for publication.
REFERENCES
- 1. Sarkozy C, Sehn LH. Management of relapsed/refractory DLBCL. Best Pract Res Clin Haematol. 2018;31:209‐216. [DOI] [PubMed] [Google Scholar]
- 2. Boslooper K, Kibbelaar R, Storm H, et al. Treatment with rituximab, cyclophosphamide, doxorubicin, vincristine and prednisolone is beneficial but toxic in very elderly patients with diffuse large B‐cell lymphoma: a population‐based cohort study on treatment, toxicity and outcome. Leuk Lymphoma. 2014;55:526‐532. [DOI] [PubMed] [Google Scholar]
- 3. Lugtenburg PJ, Lyon AR, Marks R, Luminari S. Treatment of aggressive non‐Hodgkin's lymphoma in frail patients: cardiac comorbidities and advanced age. Future Oncol. 2019;15:1197‐1205. [DOI] [PubMed] [Google Scholar]
- 4. Shi X, Liu X, Li X, et al. Risk stratification for diffuse large B‐cell lymphoma by integrating interim evaluation and International Prognostic Index: a multicenter retrospective study. Front Oncol. 2021;11:754964. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5. Chiappella A, Crombie J, Guidetti A, Vitolo U, Armand P, Corradini P. Are we ready to treat diffuse large B‐cell and high‐grade lymphoma according to major genetic subtypes? HemaSphere. 2019;3:e284. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6. Bartlett NL, Wilson WH, Jung S‐H, et al. Dose‐adjusted EPOCH‐R compared with R‐CHOP as frontline therapy for diffuse large B‐cell lymphoma: clinical outcomes of the phase III intergroup trial Alliance/CALGB 50303. J Clin Oncol. 2019;37:1790‐1799. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7. Sehn LH, Martelli M, Trněný M, et al. A randomized, open‐label, phase III study of obinutuzumab or rituximab plus CHOP in patients with previously untreated diffuse large B‐cell lymphoma: final analysis of GOYA. J Hematol Oncol. 2020;13:1‐9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8. Récher C, Coiffier B, Haioun C, et al. Intensified chemotherapy with ACVBP plus rituximab versus standard CHOP plus rituximab for the treatment of diffuse large B‐cell lymphoma (LNH03‐2B): an open‐label randomised phase 3 trial. Lancet. 2011;378:1858‐1867. [DOI] [PubMed] [Google Scholar]
- 9. Peyrade F, Jardin F, Thieblemont C, et al. Attenuated immunochemotherapy regimen (R‐miniCHOP) in elderly patients older than 80 years with diffuse large B‐cell lymphoma: a multicentre, single‐arm, phase 2 trial. Lancet Oncol. 2011;12:460‐468. [DOI] [PubMed] [Google Scholar]
- 10. Peyrade F, Bologna S, Delwail V, et al. Combination of ofatumumab and reduced‐dose CHOP for diffuse large B‐cell lymphomas in patients aged 80 years or older: an open‐label, multicentre, single‐arm, phase 2 trial from the LYSA group. Lancet Haematol. 2017;4:e46‐e55. [DOI] [PubMed] [Google Scholar]
- 11. Younes A, Sehn LH, Johnson P, et al. Randomized phase III trial of ibrutinib and rituximab plus cyclophosphamide, doxorubicin, vincristine, and prednisone in non‐germinal center B‐cell diffuse large B‐cell lymphoma. J Clin Oncol. 2019;37:1285‐1295. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12. Denker S, Bittner A, Frick M, et al. Ibrutinib‐ and bortezomib‐extended R‐CHOP induction in elderly higher‐risk patients newly diagnosed with diffuse large B‐cell lymphoma – first analysis of toxicity and efficacy signals. Leuk Lymphoma. 2022;63:84‐92. [DOI] [PubMed] [Google Scholar]
- 13. Davies AJ, Caddy J, McLaughlin K, et al. Durable responses from acalabrutinib in combination with rituximab, cyclophosphamide, doxorubicin, vincristine and prednisolone (R‐CHOP) as first line therapy for patients with diffuse large B‐cell lymphoma (DLBCL): the Accept phase Ib/II single arm study. Blood. 2022;140:9478‐9479. [Google Scholar]
- 14. Morschhauser F, Feugier P, Flinn IW, et al. A phase 2 study of venetoclax plus R‐CHOP as first‐line treatment for patients with diffuse large B‐cell lymphoma. Blood. 2021;137:600‐609. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15. Tilly H, Morschhauser F, Sehn LH, et al. Polatuzumab vedotin in previously untreated diffuse large B‐cell lymphoma. New Engl J Med. 2022;386:351‐363. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16. Nowakowski GS, Hong F, Scott DW, et al. Addition of lenalidomide to R‐CHOP improves outcomes in newly diagnosed diffuse large B‐cell lymphoma in a randomized phase II US intergroup study ECOG‐ACRIN E1412. J Clin Oncol. 2021;39:1329‐1338. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17. Nowakowski GS, Chiappella A, Gascoyne RD, et al. ROBUST: a phase III study of lenalidomide plus R‐CHOP versus placebo plus R‐CHOP in previously untreated patients with ABC‐type diffuse large B‐cell lymphoma. J Clin Oncol. 2021;39:1317‐1328. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18. Gini G, Tani M, Tucci A, et al. Lenalidomide plus rituximab for the initial treatment of frail older patients with DLBCL: the FIL_ReRi phase 2 study. Blood. 2023;142:1438‐1477. [DOI] [PubMed] [Google Scholar]
- 19. Kuzume A, Chi S, Yamauchi N, Minami Y. Immune‐checkpoint blockade therapy in lymphoma. Int J Mol Sci. 2020;21:1‐15. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20. Tamma R, Ranieri G, Ingravallo G, et al. Inflammatory cells in diffuse large B cell lymphoma. J Clin Med. 2020;9:2418. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21. Ansell SM, Stenson M, Habermann TM, Jelinek DF, Witzig TE. Cd4+ T‐cell immune response to large B‐cell non‐Hodgkin's lymphoma predicts patient outcome. J Clin Oncol. 2001;19:720‐726. [DOI] [PubMed] [Google Scholar]
- 22. Fangazio M, Ladewig E, Gomez K, et al. Genetic mechanisms of HLA‐I loss and immune escape in diffuse large B cell lymphoma. Proc Natl Acad Sci U S A. 2021;118:e2104504118. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23. Rimsza LM, Roberts RA, Miller TP, et al. Loss of MHC class II gene and protein expression in diffuse large B‐cell lymphoma is related to decreased tumor immunosurveillance and poor patient survival regardless of other prognostic factors: a follow‐up study from the Leukemia and Lymphoma Molecular. Blood. 2004;103:4251‐4258. [DOI] [PubMed] [Google Scholar]
- 24. Miller JS, Lanier LL. Natural killer cells in cancer immunotherapy. Annu Rev Cancer Biol. 2019;3:77‐103. [Google Scholar]
- 25. Gentles AJ, Newman AM, Liu CL, et al. The prognostic landscape of genes and infiltrating immune cells across human cancers. Nat Med. 2015;21:938. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26. Deng J, Fleming JB. Inflammation and myeloid cells in cancer progression and metastasis. Front Cell Dev Biol. 2022;9:759691. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27. Sterner RC, Sterner RM. CAR‐T cell therapy: current limitations and potential strategies. Blood Cancer J. 2021;11:69. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28. Jacobson C, Locke FL, Ghobadi A, et al. Long‐term (≥4 year and ≥5 year) overall survival (OS) by 12‐ and 24‐month event‐free survival (EFS): an updated analysis of ZUMA‐1, the pivotal study of axicabtagene ciloleucel (Axi‐Cel) in patients (pts) with refractory large B‐cell lymphoma (LBCL). Blood. 2021;138:1764. [Google Scholar]
- 29. Neelapu SS, Jacobson CA, Ghobadi A, et al. Five‐year follow‐up of ZUMA‐1 supports the curative potential of axicabtagene ciloleucel in refractory large B‐cell lymphoma. Blood. 2023;141:2307‐2315. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30. Schuster SJ, Tam CS, Borchmann P, et al. Long‐term clinical outcomes of tisagenlecleucel in patients with relapsed or refractory aggressive B‐cell lymphomas (JULIET): a multicentre, open‐label, single‐arm, phase 2 study. Lancet Oncol. 2021;22:1403‐1415. [DOI] [PubMed] [Google Scholar]
- 31. Abramson JS, Palomba ML, Gordon LI, et al. Lisocabtagene maraleucel for patients with relapsed or refractory large B‐cell lymphomas (TRANSCEND NHL 001): a multicentre seamless design study. Lancet. 2020;396:839‐852. [DOI] [PubMed] [Google Scholar]
- 32. Westin JR, Kersten MJ, Salles G, et al. Efficacy and safety of CD19‐directed CAR‐T cell therapies in patients with relapsed/refractory aggressive B‐cell lymphomas: observations from the JULIET, ZUMA‐1, and TRANSCEND trials. Am J Hematol. 2021;96:1295‐1312. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33. Sehgal A, Hoda D, Riedell PA, et al. Lisocabtagene maraleucel as second‐line therapy in adults with relapsed or refractory large B‐cell lymphoma who were not intended for haematopoietic stem cell transplantation (PILOT): an open‐label, phase 2 study. Lancet Oncol. 2022;23:1066‐1077. [DOI] [PubMed] [Google Scholar]
- 34. Abramson JS, Solomon SR, Arnason JE, et al. Lisocabtagene maraleucel as second‐line therapy for large B‐cell lymphoma: primary analysis of phase 3 TRANSFORM study. Blood. 2022;141:1675‐1684. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35. Locke FL, Miklos DB, Jacobson CA, et al. Axicabtagene ciloleucel as second‐line therapy for large B‐cell lymphoma. N Engl J Med. 2022;386:640‐654. [DOI] [PubMed] [Google Scholar]
- 36. Neelapu SS, Dickinson M, Munoz J, et al. Axicabtagene ciloleucel as first‐line therapy in high‐risk large B‐cell lymphoma: the phase 2 ZUMA‐12 trial. Nat Med. 2022;28:735‐742. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37. Westin J, Jacobson CA, Chavez JC, et al. ZUMA‐23: a global, phase 3, randomized controlled study of axicabtagene ciloleucel versus standard of care as first‐line therapy in patients with high‐risk large B‐cell lymphoma. J Clin Oncol. 2023;41:TPS7578. [Google Scholar]
- 38. Deng Q, Han G, Puebla‐Osorio N, et al. Characteristics of anti‐CD19 CAR T‐cell infusion products associated with efficacy and toxicity in patients with large B‐cell lymphomas. Nat Med. 2020;26:1878‐1887. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39. Zurko J, Nizamuddin I, Epperla N, 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. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40. Lee DW, Santomasso BD, Locke FL, et al. ASTCT consensus grading for cytokine release syndrome and neurologic toxicity associated with immune effector cells. Biol Blood Marrow Transplant. 2019;25:625‐638. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41. Neelapu SS, Tummala S, Kebriaei P, et al. Chimeric antigen receptor T‐cell therapy — assessment and management of toxicities. Nat Rev Clin Oncol. 2018;15:47‐62. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42. Hawkins ER, D'Souza RR, Klampatsa A. Armored CAR T‐cells: the next chapter in T‐cell cancer immunotherapy. Biol Theory. 2021;15:95‐105. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43. Yeku OO, Brentjens RJ. Armored CAR T‐cells: utilizing cytokines and pro‐inflammatory ligands to enhance CAR T‐cell anti‐tumour efficacy. Biochem Soc Trans. 2016;44:412‐418. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44. Asmamaw Dejenie T, Tiruneh G, Medhin M, et al. Current updates on generations, approvals, and clinical trials of CAR T‐cell therapy. Hum Vaccin Immunother. 2022;18:e2114254. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45. Han X, Wang Y, Wei J, Han W. Multi‐antigen‐targeted chimeric antigen receptor T cells for cancer therapy. J Hematol Oncol. 2019;12:128. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46. ClinicalTrials.gov . Study of IMPT‐314 in R/R aggressive B‐cell NHL [Internet]. Accessed September 2, 2024. https://www.clinicaltrials.gov/study/NCT05826535
- 47. Depil S, Duchateau P, Grupp SA, Mufti G, Poirot L. “Off‐the‐shelf” allogeneic CAR T cells: development and challenges. Nat Rev Drug Discov. 2020;19:185‐199. [DOI] [PubMed] [Google Scholar]
- 48. Khurana A, Lin Y. Allogeneic chimeric antigen receptor therapy in lymphoma. Curr Treat Options Oncol. 2022;23:171‐187. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49. Locke FL, Malik S, Tees MT, et al. First‐in‐human data of ALLO‐501A, an allogeneic chimeric antigen receptor (CAR) T‐cell therapy and ALLO‐647 in relapsed/refractory large B‐cell lymphoma (R/R LBCL): ALPHA2 study. J Clin Oncol. 2021;39:2529. [Google Scholar]
- 50. Salvaris R, Ong J, Gregory GP. Bispecific antibodies: a review of development, clinical efficacy and toxicity in B‐cell lymphomas. J Pers Med. 2021;11:355. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51. Falchi L, Vardhana SA, Salles GA. Bispecific antibodies for the treatment of B‐cell lymphoma: promises, unknowns, and opportunities. Blood. 2023;141:467‐480. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52. Sehn LH, Chamuleau M, Lenz G, et al. Phase 3 trial of subcutaneous epcoritamab + R‐CHOP versus R‐CHOP in patients (pts) with newly diagnosed diffuse large B‐cell lymphoma (DLBCL): EPCORE DLBCL‐2. Hematol Oncol. 2023;41:849‐850. [Google Scholar]
- 53. ClinicalTrials.gov . A study of TNB‐486 in subjects with relapsed or refractory B‐cell non‐Hodgkin lymphoma [Internet]; 2020. Accessed May 19, 2023. https://clinicaltrials.gov/ct2/show/NCT04594642
- 54. González BE. Role of bispecific antibodies in relapsed/refractory diffuse large B‐cell lymphoma in the CART era. Front Immunol. 2022;13:909008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55. Lu H, Oka A, Coulson M, et al. PIT565, a first‐in‐class anti‐CD19, anti‐CD3, anti‐CD2 trispecific antibody for the treatment of B cell malignancies. Blood. 2022;140:3148. [Google Scholar]
- 56. ClinicalTrials.gov . A study of JNJ‐80948543, a T‐cell redirecting CD79b x CD20 x CD3 trispecific antibody, in participants with non‐Hodgkin lymphoma (NHL) and chronic lymphocytic leukemia (CLL) [Internet]. Accessed March 13, 2024. https://www.clinicaltrials.gov/study/NCT05424822
- 57. Dickinson MJ, Carlo‐Stella C, Morschhauser F, et al. Glofitamab for relapsed or refractory diffuse large B‐cell lymphoma. N Engl J Med. 2022;387:2220‐2231. [DOI] [PubMed] [Google Scholar]
- 58. Abramson JS, Palomba ML, Gordon LI, et al. Two‐year follow‐up of Transcend NHL 001, a multicenter phase 1 study of lisocabtagene maraleucel (liso‐cel) in relapsed or refractory (R/R) large B‐cell lymphomas (LBCL). Blood. 2021;138:2840. [DOI] [PubMed] [Google Scholar]
- 59. Johnson NA, Boyle M, Bashashati A, et al. Diffuse large B‐cell lymphoma: reduced CD20 expression is associated with an inferior survival. Blood. 2009;113:3773‐3780. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60. Prevodnik VK, Lavrenčak J, Horvat M, Novakovič BJ. The predictive significance of CD20 expression in B‐cell lymphomas. Diagn Pathol. 2011;6:33. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61. Horton HM, Bernett MJ, Pong E, et al. Potent in vitro and in vivo activity of an Fc‐engineered anti‐CD19 monoclonal antibody against lymphoma and leukemia. Cancer Res. 2008;68:8049‐8057. [DOI] [PubMed] [Google Scholar]
- 62. Katz BZ, Herishanu Y. Therapeutic targeting of CD19 in hematological malignancies: past, present, future and beyond. Leuk Lymphoma. 2014;55:999‐1006. [DOI] [PubMed] [Google Scholar]
- 63. Salles G, Długosz‐Danecka M, Ghesquières H, Jurczak W. Tafasitamab for the treatment of relapsed or refractory diffuse large B‐cell lymphoma. Expert Opin Biol Ther. 2021;21:455‐463. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64. Her JH, Pretscher D, Patra‐Kneuer M, et al. Tafasitamab mediates killing of B‐cell non‐Hodgkin's lymphoma in combination with γδ T cell or allogeneic NK cell therapy. Cancer Immunol Immunother. 2022;71:2829‐2836. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65. Awan FT, Lapalombella R, Trotta R, et al. CD19 targeting of chronic lymphocytic leukemia with a novel Fc‐domain‐engineered monoclonal antibody. Blood. 2010;115:1204‐1213. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66. Duell J, Maddocks KJ, González‐Barca E, et al. Long‐term outcomes from the phase II L‐MIND study of tafasitamab (MOR208) plus lenalidomide in patients with relapsed or refractory diffuse large B‐cell lymphoma. Haematologica. 2021;106:2417‐2426. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67. Duell J, Abrisqueta P, Andre M, et al. Tafasitamab for patients with relapsed or refractory diffuse large B‐cell lymphoma: final 5‐year efficacy and safety findings in the phase II L‐MIND study. Haematologica. 2024;109:553‐566. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68. Chu Y, Lamb M, Cairo MS, Lee DA. The future of natural killer cell immunotherapy for B cell non‐Hodgkin lymphoma (B cell NHL). Curr Treat Options Oncol. 2022;23:381‐403. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69. Liu E, Marin D, Banerjee P, et al. Use of CAR‐transduced natural killer cells in CD19‐positive lymphoid tumors. N Engl J Med. 2020;382:545‐553. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70. Xie G, Dong H, Liang Y, Ham JD, Rizwan R, Chen J. CAR‐NK cells: a promising cellular immunotherapy for cancer. EBioMedicine. 2020;59:102975. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71. Sabbah M, Jondreville L, Lacan C, et al. CAR‐NK cells: a chimeric hope or a promising therapy? Cancers (Basel). 2022;14:3839. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72. Nieto Y, Banerjee PP, Kaur I, et al. Innate cell engager AFM13 combined with preactivated and expanded cord blood‐derived NK cells for patients with double refractory CD30+ lymphoma. Blood. 2022;140:415‐416.35925646 [Google Scholar]
- 73. Eladl E, Tremblay‐Lemay R, Rastgoo N, et al. Role of CD47 in hematological malignancies. J Hematol Oncol. 2020;13:1‐14. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74. Tun AM, Ansell SM. Immunotherapy in Hodgkin and non‐Hodgkin lymphoma: innate, adaptive and targeted immunological strategies. Cancer Treat Rev. 2020;88:102042. [DOI] [PubMed] [Google Scholar]
- 75. Chao MP, Alizadeh AA, Tang C, et al. Anti‐CD47 antibody synergizes with rituximab to promote phagocytosis and eradicate non‐Hodgkin lymphoma. Cell. 2010;142:699‐713. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76. Klichinsky M, Ruella M, Shestova O, et al. Human chimeric antigen receptor macrophages for cancer immunotherapy. Nat Biotechnol. 2020;38:947‐953. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77. Kennedy GA, Tey SK, Cobcroft R, et al. Incidence and nature of CD20‐negative relapses following rituximab therapy in aggressive B‐cell non‐Hodgkin's lymphoma: a retrospective review. Br J Haematol. 2002;119:412‐416. [DOI] [PubMed] [Google Scholar]
- 78. Belada D, Kopeckova K, Bergua Burgues JM, et al. Safety and efficacy of tafasitamab ± lenalidomide added to first‐line R‐CHOP for DLBCL: phase 1b first‐MIND study. Blood. 2023;142:1348‐1358. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79. ClinicalTrials.gov . A multicentre, parallel arm, open‐label trial of frontline R‐CHOP/pola‐RCHP and glofitamab in younger, higher risk patients with diffuse large B cell lymphoma (DLBCL) (COALITION) [Internet]; 2021. Accessed. April 19; 2023. https://clinicaltrials.gov/ct2/show/NCT04914741
- 80. Sehn LH, Hertzberg M, Opat S, et al. Polatuzumab vedotin plus bendamustine and rituximab in relapsed/refractory DLBCL: survival update and new extension cohort data. Blood Adv. 2022;6:533‐543. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81. Westin J, Olszewski AJ, Fogliatto L, et al. SUNMO: a phase III trial evaluating the efficacy and safety of mosunetuzumab in combination with polatuzumab vedotin vs rituximab plus gemcitabine and oxaliplatin in patients with relapsed/refractory aggressive B‐cell non‐Hodgkin lymphoma. J Clin Oncol. 2023;41:TPS7586. [Google Scholar]
- 82. Nakamura A, Grossman S, Song K, et al. The SUMOylation inhibitor subasumstat potentiates rituximab activity by IFN1‐dependent macrophage and NK cell stimulation. Blood. 2022;139:2770‐2781. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83. ClinicalTrials.gov . Subcutaneous epcoritamab with or without lenalidomide as first line therapy for diffuse large B‐cell lymphoma [Internet]; 2022. Accessed April 19, 2023. https://clinicaltrials.gov/ct2/show/NCT05660967
- 84. Westin JR, Steiner R, Feng L, et al. Smart Stop: a phase II study of lenalidomide, tafasitamab, rituximab, and acalabrutinib alone and with chemotherapy in patients with newly diagnosed DLBCL. Blood. 2021;138:3572. [Google Scholar]
- 85. Westin J, Davis RE, Feng L, et al. Smart Start: rituximab, lenalidomide, and ibrutinib in patients with newly diagnosed large B‐cell lymphoma. J Clin Oncol. 2023;41:745‐755. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86. Westin J, Burke JM, Chapman AE, et al. A phase 2, open‐label study of loncastuximab tesirine in combination with rituximab (lonca‐R) in previously untreated unfit/frail patients with diffuse large B‐cell lymphoma (DLBCL) (LOTIS‐9). Blood. 2022;140:6638‐6639. [Google Scholar]
- 87. ClinicalTrials.gov . A phase II trial of mosunetuzumab, polatuzumab, tafasitamab, and lenalidomide in patients with relapsed B‐cell NHL [Internet]; 2022. Accessed May 19, 2023. https://clinicaltrials.gov/ct2/show/NCT05615636
- 88. Sakemura R, Horvei P, Manriquez‐Roman C, et al. Poster 2412: the impact of prior treatment with a CD19 targeting monoclonal antibody on subsequent treatment with CD19 targeting CART cell therapy in preclinical models. Blood. 2021;138:2412. [Google Scholar]
- 89. Sakemura RL, Manriquez‐Roman C, Horvei P, et al. CD19 antigen occupancy on cancer cells with the CD19 monoclonal antibody tafasitamab improves the activation, antitumor efficacy, and safety profile of CART19 cell therapy. Blood. 2022;140:2362‐2364. [Google Scholar]
- 90. ADC Therapeutics SA . ADC Therapeutics announces plan to discontinue the phase 2 LOTIS‐9 clinical trial of ZYNLONTA® (loncastuximab tesirine‐lpyl) and rituximab in unfit or frail previously untreated DLBCL patients [Internet]. Accessed February 9, 2024. https://ir.adctherapeutics.com/press‐releases/press‐release‐details/2023/ADC‐Therapeutics‐Announces‐Plan‐to‐Discontinue‐the‐Phase‐2‐LOTIS‐9‐Clinical‐Trial‐of‐ZYNLONTA‐loncastuximab‐tesirine‐lpyl‐and‐Rituximab‐in‐Unfit‐or‐Frail‐Previously‐Untreated‐DLBCL‐P
- 91. ADC Therapeutics SA . ADC Therapeutics announces voluntary pause of enrollment in the phase 2 LOTIS‐9 clinical trial of ZYNLONTA® (loncastuximab tesirine‐lpyl) and rituximab in unfit or frail previously untreated DLBCL patients [Internet]. Accessed February 9, 2024. https://ir.adctherapeutics.com/press‐releases/press‐release‐details/2023/ADC‐Therapeutics‐Announces‐Voluntary‐Pause‐of‐Enrollment‐in‐the‐Phase‐2‐LOTIS‐9‐Clinical‐Trial‐of‐ZYNLONTA‐loncastuximab‐tesirine‐lpyl‐and‐Rituximab‐in‐Unfit‐or‐Frail‐Previously‐Untr
- 92. Chavez JC, Nastoupil LJ, Morschhauser F, et al. Efficacy and safety of golcadomide, a novel cereblon E3 ligase modulator (CELMoD) agent, combined with rituximab in a phase 1/2 open‐label study of patients with relapsed/refractory non‐Hodgkin lymphoma. Blood. 2023;142:4496. [Google Scholar]
- 93. Nastoupil LJ, Kuruvilla J, Chavez JC, et al. Phase Ib study of avadomide (CC‐122) in combination with rituximab in patients with relapsed/refractory diffuse large B‐cell lymphoma and follicular lymphoma. EJHaem. 2022;3:394‐405. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94. Davoodi‐Moghaddam Z, Jafari‐Raddani F, Noori M, Bashash D. A systematic review and meta‐analysis of immune checkpoint therapy in relapsed or refractory non‐Hodgkin lymphoma; a friend or foe? Transl Oncol. 2023;30:101636. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95. ClinicalTrials.gov . A phase I clinical study of HLX53 in advanced/metastatic solid tumors or lymphoma [Internet]; 2022. Accessed May 19, 2023. https://clinicaltrials.gov/ct2/show/NCT05394168
- 96. ClinicalTrials.gov . Efficacy and safety of IBI110 single agent and in combination with sintilimab in patients with relapsed or refractory diffuse large B cell lymphoma (r/r DLBCL) [Internet]; 2022. Accessed May 19, 2023. https://clinicaltrials.gov/ct2/show/NCT05039658
- 97. ClinicalTrials.gov . A safety and tolerability study of INCAGN02385 in select advanced malignancies [Internet]; 2020. Accessed May 19, 2023. https://clinicaltrials.gov/ct2/show/NCT03538028
- 98. ClinicalTrials.gov . Sym023 (anti‐TIM‐3) in patients with advanced solid tumor malignancies or lymphomas [Internet]; 2021. Accessed May 19, 2023. https://clinicaltrials.gov/ct2/show/NCT03489343
- 99. ClinicalTrials.gov . To evaluate the safety and efficacy of TQB2618 injection combined with penpulimab in the treatment of patients with relapsed and refractory lymphoma [Internet]; 2023. Accessed May 19, 2023. https://clinicaltrials.gov/ct2/show/NCT05400876
- 100. ClinicalTrials.gov . A study to evaluate the safety, tolerability, and efficacy of relatlimab in relapsed or refractory B‐cell malignancies [Internet]; 2023. Accessed May 19, 2023. https://www.clinicaltrials.gov/ct2/show/NCT02061761
- 101. Advani R, Flinn I, Popplewell L, et al. CD47 blockade by Hu5F9‐G4 and rituximab in non‐Hodgkin's lymphoma. N Engl J Med. 2018;379:1711‐1721. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102. Advani R, Bartlett NL, Smith SM, et al. The first‐in‐class anti‐CD47 antibody HU5F9‐G4 + rituximab induces durable responses in relapsed/refractory DLBCL and indolent lymphoma: interim phase 1B/2 results. Hematol Oncol. 2019;37:89‐90. [Google Scholar]
- 103. Mangelberger D, Augsberger C, Landgraf K, Heitmüller C, Steidl S. Blockade of the CD47/SIRPα checkpoint potentiates the anti‐tumor efficacy of tafasitamab. Blood. 2020;136:11‐12.32276273 [Google Scholar]
- 104. Biedermann A, Mangelberger‐Eberl D, Mougiakakos D, et al. Blocking the CD47‐Sirpa axis enhances tafasitamab‐mediated phagocytosis. Blood. 2022;140:9292‐9293. [Google Scholar]
- 105. Ng WL, Ansell SM, Mondello P. Insights into the tumor microenvironment of B cell lymphoma. J Exp Clin Cancer Res. 2022;41:362. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106. Kline J, Godfrey J, Ansell SM. The immune landscape and response to immune checkpoint blockade therapy in lymphoma. Blood. 2020;135:523‐533. [DOI] [PubMed] [Google Scholar]
- 107. Liu Y, Zhou X, Wang X. Targeting the tumor microenvironment in B‐cell lymphoma: challenges and opportunities. J Hematol Oncol. 2021;14:125. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108. Ennishi D, Jiang A, Boyle M, et al. Double‐hit gene expression signature defines a distinct subgroup of germinal center B‐cell‐like diffuse large B‐cell lymphoma. J Clin Oncol. 2019;37:190‐201. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109. Vivier E, Raulet DH, Moretta A, et al. Innate or adaptive immunity? The example of natural killer cells. Science. 2011;331:44‐49. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110. Liu S, Nguyen K, Park D, Wong N, Wang A, Zhou Y. Harnessing natural killer cells to develop next‐generation cellular immunotherapy. Chronic Dis Transl Med. 2022;8:245‐255. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111. Gumber D, Wang LD. Improving CAR‐T immunotherapy: overcoming the challenges of T cell exhaustion. EBioMedicine. 2022;77:103941. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112. Wherry E. T Cell exhaustion. Nat Immunol. 2011;12:492‐499. [DOI] [PubMed] [Google Scholar]
- 113. Chen BJ, Dashnamoorthy R, Galera P, et al. The immune checkpoint molecules PD‐1, PD‐L1, TIM‐3 and LAG‐3 in diffuse large B‐cell lymphoma. Oncotarget. 2019;10:2030‐2040. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114. Josefsson SE, Beiske K, Blaker YN, et al. TIGIT and PD‐1 mark Intratumoral T cells with reduced effector function in B‐cell non‐Hodgkin lymphoma. Cancer Immunol Res. 2019;7:355‐362. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115. Catakovic K, Klieser E, Neureiter D, Geisberger R. T cell exhaustion: from pathophysiological basics to tumor immunotherapy. Cell Commun Signal. 2017;15:1‐16. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116. Chandrasekaran S, Funk CR, Kleber T, Paulos CM, Shanmugam M, Waller EK. Strategies to overcome failures in T‐cell immunotherapies by targeting PI3K‐δ and ‐γ. Front Immunol. 2021;12:718621. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 117. Profitós‐Pelejà N, Santos JC, Marín‐Niebla A, Roué G, Ribeiro ML. Regulation of B‐cell receptor signaling and its therapeutic relevance in aggressive B‐cell lymphomas. Cancers (Basel). 2022;14:860. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118. Morin RD, Arthur SE, Hodson DJ. Molecular profiling in diffuse large B‐cell lymphoma: why so many types of subtypes? Br J Haematol. 2022;196:814‐829. [DOI] [PubMed] [Google Scholar]
- 119. Mondello P, Ansell SM, Nowakowski GS. Immune epigenetic crosstalk between malignant B cells and the tumor microenvironment in B cell lymphoma. Front Genet. 2022;13:826594. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120. Ennishi D, Takata K, Béguelin W, et al. Molecular and genetic characterization of MHC deficiency identifies ezh2 as therapeutic target for enhancing immune recognition. Cancer Discov. 2019;9:546‐563. [DOI] [PubMed] [Google Scholar]
- 121. Hu M, Li Y, Lu Y, et al. The regulation of immune checkpoints by the hypoxic tumor microenvironment. Peer J. 2021;9:e11306. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122. Henze AT, Mazzone M. The impact of hypoxia on tumor‐associated macrophages. J Clin Invest. 2016;126:3672‐3679. [DOI] [PMC free article] [PubMed] [Google Scholar]
