Several second- and third-line immunotherapeutic options for patients with relapsed or refractory diffuse large B-cell lymphoma ineligible for autologous stem cell transplant are directed against the B-cell antigen cluster of differentiation 19 (CD19). The anti–CD19 monoclonal antibody tafasitamab, paired with the immunomodulator lenalidomide, mediates antibody-dependent cellular toxicity and phagocytosis; the antibody–drug conjugate loncastuximab tesirine delivers the DNA cross-linking agent tesirine via CD19 binding and internalization; and CD19-directed chimeric antigen receptor T-cell therapy (CAR-T) products are engineered from autologous T cells. Although CD19 expression is assessed at diagnosis, clinically relevant thresholds of CD19 expression—which may not be detectable using current routine methodologies—have not been defined and may vary between CD19-directed treatment modalities. Determining optimal treatment sequencing strategies for CD19-directed therapy is hampered by the exclusion of patients who have received prior CD19-directed therapies from major clinical trials. Antigen escape, which is attributed to mechanisms including epitope loss and defective cell surface trafficking of CD19, is an important cause of CAR-T failure. Limited data suggest that CD19 expression may be maintained after non–CAR-T CD19-directed therapy, and retrospective analyses indicate that some patients with disease relapse after CAR-T may benefit from subsequent CD19-directed therapy. To date, clinical evidence on the effect of anti-CD19 therapy prior to CAR-T has been limited to small case series. Prospective studies and detailed analyses are needed to understand how pretreatment and posttreatment CD19 expression correlates with clinical responses to subsequent CD19-directed therapy to fully maximize treatment strategies.

As immunotherapies have expanded the range of treatment options for diffuse large B-cell lymphoma (DLBCL), the complexity of optimal treatment sequencing has concomitantly increased. Frontline treatment is composed of anthracycline-based combination chemotherapy combined with anti–cluster of differentiation 20 (CD20) monoclonal antibodies, most commonly rituximab, cyclophosphamide, doxorubicin, vincristine, and prednisone (R-CHOP; refs. 16). Although this approach can cure a significant subset of patients, approximately 40% of patients have a disease that fails to respond or subsequently relapses (7). Second-line therapy is informed by the response to frontline treatment. For patients with refractory or early relapsed (<12 months) disease, CD19-directed chimeric antigen receptor (CAR) T-cell therapy (CAR-T) can offer clinical benefits, whereas disease relapse beyond 1 year is managed with platinum-based chemotherapy followed by autologous stem cell transplant (ASCT) in the event of disease response (8). However, many patients are ineligible for CAR-T or ASCT due to comorbidities or logistics or do not respond well to CAR-T or ASCT.

Whatever the reason for looking beyond CAR-T and ASCT, recently approved therapies have broadened the therapeutic armamentarium for these patients to include immunotherapy regimens such as tafasitamab plus lenalidomide, polatuzumab vedotin combined with bendamustine plus rituximab, and loncastuximab tesirine (8). Bispecific antibody (BsAb) is an emerging therapeutic option for patients with relapsed/refractory (R/R) DLBCL, including the recently approved CD20xCD3-directed antibodies glofitamab and epcoritamab (9, 10) and odronextramab, which is currently under regulatory review (11), with CD19xCD3-targeting bispecifics also in development (12). However, response data are currently substantially more mature for CD19-based treatments (1315).

Many of the recently introduced novel second- and third-line immunotherapies are directed against CD19, a B-cell–specific cluster of differentiation molecules expressed on the surface of native and malignant B cells (16). Unlike CD20, CD19 is expressed throughout all B-cell maturation stages (17), is more homogenous, and is commonly preserved in CD20-negative tumor subsets (18, 19). Data on the potential effect of previous CD19-directed treatment before the initiation of CAR-T are limited, and the optimal sequence of CD19-directed therapies is unclear. CD19-directed therapies are under evaluation in the frontline setting, including the phase III frontMIND study (NCT04824092), which evaluated tafasitamab plus lenalidomide with R-CHOP in high-intermediate– and high-risk DLBCL (20). This suggests that the future therapeutic landscape for DLBCL may include CD19-directed immunotherapies in an earlier line of treatment. Understanding ways to optimally sequence these agents is becoming increasingly relevant.

Although several new B-cell targets for lymphoma-directed therapy have emerged, in this review, we examine potential sequencing considerations for CD19-directed treatment in patients with R/R DLBCL, the effect of CD19-directed therapy on the dynamics of subsequent CD19 expression in malignant cells, the effect of CD19-directed treatment on subsequent CAR-T outcomes, and patient-related factors that may influence CD19-directed treatment selection.

Currently available CD19-directed therapies for the treatment of R/R DLBCL include tafasitamab, loncastuximab tesirine, and CAR-T products [axicabtagene ciloleucel (axi-cel), tisagenlecleucel (tisa-cel), and lisocabtagene maraleucel (liso-cel)], all of which have different mechanisms of action (Fig. 1; Table 1).

Figure 1.

Mechanisms of action in lymphoma. A, CD19 signaling pathway. B, Interaction of treatments with a lymphoma cell. ADCC, antibody-dependent cellular cytotoxicity; ADCP, antibody-dependent cellular phagocytosis; NK, natural killer; PBD, pyrrolobenzodiazepine.

Figure 1.

Mechanisms of action in lymphoma. A, CD19 signaling pathway. B, Interaction of treatments with a lymphoma cell. ADCC, antibody-dependent cellular cytotoxicity; ADCP, antibody-dependent cellular phagocytosis; NK, natural killer; PBD, pyrrolobenzodiazepine.

Close modal
Table 1.

Summary of approved CD19-directed therapies for R/R DLBCL.a

RegimenMechanism of actionTrialTrial designCancer types (n)Median follow-up, monthsORR, %CRR, %Median PFS or EFS, monthsMedian DOR in patients with response, monthsMedian OS, months
Axicabtagene ciloleucel Anti-CD19 CAR-T + CD28 CS domain ZUMA-1 (33Phase I/II DLBCL (77), PMBCL (8), and TFL (16) 15.4 82 54 5.8 (PFS) NR NR 
ZUMA-1, extended follow-up (110Phase I/II, extended follow-up DLBCL (77), PMBCL (8), and TFL (16) 63.1 83 58 5.9 (PFS) 62.2 25.8 
ZUMA-7 (35Phase III, axi-cel vs. SOC (chemoimmunotherapy followed by ASCT) as second-line therapy for R/R LBCL DLBCL (126), HGBCL (31), not confirmed (18), and other (5) 24.9 83 65 14.7 (PFS) and 8.3 (EFS) 26.9 NR 
ZUMA-7, extended follow-up (14Phase III, axi-cel vs. SOC (chemotherapy + ASCT) as second-line therapy for R/R LBCL, extended follow-up DLBCL (126), HGBCL (31), not confirmed (18), and other (5) 47.2 83 61 14.7 (PFS) and 10.8 (EFS) 41.7 NR 
Tisagenlecleucel Anti-CD19 CAR-T + 4-1BB CS domain JULIET (29Phase II DLBCL (88), TFL (21), and other (2) 14 52 40 Not reported NR 8.3 
JULIET, extended follow-up (15Phase II, extended follow-up DLBCL (92), HGBCL (17), and TFL (21) 40.3 53 39 2.9 (PFS) and 2.8 (EFS) Not estimable 11.1 
BELINDA (36Phase III, tisa-cel vs. SOC as second-line therapy for R/R aggressive B-cell lymphoma DLBCL (101), HGBCL (39), PMBCL (12) FL (5), and other (5) 10 46.30 28.40 3 (EFS) Not reported Not estimable 
Lisocabtagene maraleucel Anti-CD19 CAR-T + 4-1BB CS domain TRANSCEND NHL 001 (37Phase I/II DLBCL (215), HGBCL (36), PMBCL (15) and FL (3) 18.8 73 53 6.8 (PFS) NR 21.1 
TRANSCEND, extended follow-up (38Phase I/II, extended follow-up DLBCL (276), HGBCL (44), PMBCL (21), and FL (4) 24 73 53 6.8 (PFS) 26.1 27.3 
TRANSFORM (39Phase III, liso-cel vs. SOC (chemotherapy + ASCT) as second-line therapy for R/R LBCL, extended follow-up DLBCL (66), HGBCL (22), PMBCL (8), THRBCL (1), and FL (1) 17.5 87 74 NR (EFS and PFS) NR 29.9 
Tafasitamab + lenalidomide Anti–CD19-induced ADCC and ADCP L-MIND (23Phase II DLBCL (81) 13.2 60 43 12.1 (PFS) 21.7 NR 
L-MIND, extended follow-up (13Phase II, extended follow-up DLBCL (81) 44 57.50 41.20 11.6 (PFS) NR 33.5 
Loncastuximab tesirine Anti-CD19 ADC LOTIS-2 (26Phase II DLBCL (127), HGBCL (11), and PMBCL (7) 7.8 48.30 24 4.9 (PFS) 10.3 9.9 
LOTIS-2, extended follow-up (27Phase II, extended follow-up DLBCL (127), HGBCL (11), and PMBCL (7) 7.8 48 25 4.9 (PFS) NR 9.5 
RegimenMechanism of actionTrialTrial designCancer types (n)Median follow-up, monthsORR, %CRR, %Median PFS or EFS, monthsMedian DOR in patients with response, monthsMedian OS, months
Axicabtagene ciloleucel Anti-CD19 CAR-T + CD28 CS domain ZUMA-1 (33Phase I/II DLBCL (77), PMBCL (8), and TFL (16) 15.4 82 54 5.8 (PFS) NR NR 
ZUMA-1, extended follow-up (110Phase I/II, extended follow-up DLBCL (77), PMBCL (8), and TFL (16) 63.1 83 58 5.9 (PFS) 62.2 25.8 
ZUMA-7 (35Phase III, axi-cel vs. SOC (chemoimmunotherapy followed by ASCT) as second-line therapy for R/R LBCL DLBCL (126), HGBCL (31), not confirmed (18), and other (5) 24.9 83 65 14.7 (PFS) and 8.3 (EFS) 26.9 NR 
ZUMA-7, extended follow-up (14Phase III, axi-cel vs. SOC (chemotherapy + ASCT) as second-line therapy for R/R LBCL, extended follow-up DLBCL (126), HGBCL (31), not confirmed (18), and other (5) 47.2 83 61 14.7 (PFS) and 10.8 (EFS) 41.7 NR 
Tisagenlecleucel Anti-CD19 CAR-T + 4-1BB CS domain JULIET (29Phase II DLBCL (88), TFL (21), and other (2) 14 52 40 Not reported NR 8.3 
JULIET, extended follow-up (15Phase II, extended follow-up DLBCL (92), HGBCL (17), and TFL (21) 40.3 53 39 2.9 (PFS) and 2.8 (EFS) Not estimable 11.1 
BELINDA (36Phase III, tisa-cel vs. SOC as second-line therapy for R/R aggressive B-cell lymphoma DLBCL (101), HGBCL (39), PMBCL (12) FL (5), and other (5) 10 46.30 28.40 3 (EFS) Not reported Not estimable 
Lisocabtagene maraleucel Anti-CD19 CAR-T + 4-1BB CS domain TRANSCEND NHL 001 (37Phase I/II DLBCL (215), HGBCL (36), PMBCL (15) and FL (3) 18.8 73 53 6.8 (PFS) NR 21.1 
TRANSCEND, extended follow-up (38Phase I/II, extended follow-up DLBCL (276), HGBCL (44), PMBCL (21), and FL (4) 24 73 53 6.8 (PFS) 26.1 27.3 
TRANSFORM (39Phase III, liso-cel vs. SOC (chemotherapy + ASCT) as second-line therapy for R/R LBCL, extended follow-up DLBCL (66), HGBCL (22), PMBCL (8), THRBCL (1), and FL (1) 17.5 87 74 NR (EFS and PFS) NR 29.9 
Tafasitamab + lenalidomide Anti–CD19-induced ADCC and ADCP L-MIND (23Phase II DLBCL (81) 13.2 60 43 12.1 (PFS) 21.7 NR 
L-MIND, extended follow-up (13Phase II, extended follow-up DLBCL (81) 44 57.50 41.20 11.6 (PFS) NR 33.5 
Loncastuximab tesirine Anti-CD19 ADC LOTIS-2 (26Phase II DLBCL (127), HGBCL (11), and PMBCL (7) 7.8 48.30 24 4.9 (PFS) 10.3 9.9 
LOTIS-2, extended follow-up (27Phase II, extended follow-up DLBCL (127), HGBCL (11), and PMBCL (7) 7.8 48 25 4.9 (PFS) NR 9.5 

ADCC, antibody-dependent cell-mediated cytotoxicity; ADCP, antibody-dependent cellular phagocytosis; CS, costimulatory; DOR, duration of response; FL, follicular lymphoma; HGBCL, high-grade B-cell lymphoma; NR, not reached; PMBCL, primary mediastinal large B-cell lymphoma; TFL, transformed follicular lymphoma; THRBCL, T-cell/histiocyte-rich large B-cell lymphoma.

a

This summary is provided for ease of reference to the applicable studies only. The populations and methodology are substantially different in each study, and outcomes should not be directly compared.

Tafasitamab is an Fc-enhanced, humanized, anti–CD19 monoclonal antibody that functions as an immunotherapeutic agent by mediating antibody-dependent cellular toxicity and antibody-dependent cellular phagocytosis (21). Tafasitamab is administered in combination with the immunomodulator lenalidomide (21), which has antineoplastic, antiangiogenic, and proerythropoietic properties (22). In the phase II L-MIND study, patients receiving tafasitamab achieved an overall response rate (ORR), complete response (CR) rate (CRR), and a median progression-free survival (mPFS) of 60%, 43%, and 12.1 months, respectively (23). The updated 5-year analysis showed similar ORR, CRR, and mPFS but demonstrated a median overall survival (mOS) of 33.5 months (13).

Loncastuximab tesirine is an anti-CD19 antibody–drug conjugate (ADC) that delivers the conjugated drug tesirine, a chemotherapeutic agent that cross-links DNA in target cells, after binding and internalization (24, 25). When evaluated in the phase II LOTIS-2 trial, loncastuximab tesirine showed an ORR of 48.3%, with 24% of patients experiencing CR, and an mOS of 9.5 months (26, 27).

CAR-T products are derived from genetically modified autologous T cells transduced by ex vivo lentiviral- or adeno-associated viral transgene insertion to express a CAR specific for CD19 (2831). This process takes 8 to 37 days (28, 30, 32), depending on the specific CAR-T–cell product used (30). Bridging therapy is commonly administered during the CAR-T–cell manufacturing process (8).

Axi-cel is an anti-CD19 CAR-T with a CD28 costimulatory domain. The phase II ZUMA-1 trial demonstrated, after 5 years of follow-up, an ORR with axi-cel, a CRR, a mOS, and a median duration of CR of 83%, 58%, 25.8 months, and 62.2 months, respectively (33, 34). The phase III ZUMA-7 trial that compared axi-cel with standard of care (SOC) chemotherapy followed by ASCT in patients with R/R DLBCL showed improved event-free survival (EFS) in patients receiving axi-cel versus SOC (8.3 vs. 2 months), mPFS (14.7 vs. 3.7 months), and mOS (not reached vs. 31.1 months; ref. 35).

Tisa-cel is an anti-CD19 CAR-T with a 4-1BB costimulatory domain. In the phase II JULIET study, tisa-cel demonstrated an ORR, a CRR, and an mOS of 52%, 40%, and 8.3 months, respectively (29), with similar results in the long-term JULIET analysis (15). In BELINDA, a phase III trial, tisa-cel was found not to be superior to SOC, with both groups showing an mEFS of 3 months (36).

Liso-cel is an anti-CD19 CAR-T with a 4-1BB costimulatory domain, which, in the phase II TRANSCEND trial, achieved an ORR, a CRR, and a mPFS of 73%, 53%, and 6.8 months, respectively (37). An updated analysis of the BELINDA study at 2 year follow-up showed similar response rates, a median response duration of 26.1 months, and an mOS of 27.3 months (38). In the phase III TRANSFORM study, liso-cel, as compared with SOC, showed improvements in EFS (not reached vs. 2.4 months), CRR (74% vs. 34%), mOS (not reached vs. 29.9 months), and mPFS (not reached vs. 6.2 months; ref. 39).

All three randomized trials conducted to compare the effect of CAR-T on salvage chemotherapy followed by ASCT in patients with primary refractory or early relapsing large B-cell lymphoma (LBCL; ZUMA-7, BELINDA, and TRANSFORM) had similar objectives but yielded discordant results (35, 36, 39). Acknowledging the limitations of cross-trial comparisons, differences in trial design, definitions of EFS, CAR-T manufacturing timelines, and participant heterogeneity may have contributed to these discrepancies. The BELINDA and TRANSFORM studies permitted bridging chemotherapy, whereas the ZUMA-7 study permitted only steroids, potentially introducing selection bias for less aggressive lymphoma in the ZUMA-7 cohort. In the two studies that permitted crossover, BELINDA and TRANSFORM, the former allowed crossover only after progressive disease following two lines of salvage therapy, whereas the latter required progressive disease after only one line of salvage chemotherapy, potentially introducing a selection bias of more heavily pretreated patients in the CAR-T arm of BELINDA. Regarding the intertrial variability in defining EFS, the BELINDA study was the only trial that did not include the initiation of new lymphoma therapy as an event, and all three trials assigned a different response assessment time point as event-defining. Although the TRANSFORM study did not report the median time from randomization to CAR-T infusion, there was a marked difference in the ZUMA-7 study compared with the BELINDA study (median, 29 vs. 52 days, respectively). The CAR-T manufacturing challenges in the BELINDA study may have led to participants with higher tumor burden and declining functional status at the time of CAR-T infusion, two clinical characteristics associated with lower CAR-T efficacy (4042).

Understanding the effects of CD19-directed therapeutic modalities on CD19 expression is important for developing rational treatment strategies across therapeutic lines. However, the level of tumor cell CD19 expression required for the clinical response to CD19-directed therapy is unclear and may differ among drugs. Furthermore, data on clinical activity in R/R DLBCL using sequential CD19-directed therapies are limited because several pivotal trials leading to regulatory approval excluded patients who had received prior CD19-directed therapy (23, 29, 33, 35, 43).

CD19 detection and expression level assessment

DLBCL treatment guidelines recommend testing for CD19 expression as part of the diagnostic process (8, 44). The two methods routinely used in clinical practice to assess CD19 include immunohistochemistry (IHC) and flow cytometry. Although valuable for initial screening, the applicability of these methods for monitoring CD19 expression after CD19-directed therapy and guiding subsequent treatment strategies is unclear (4547).

A small, growing body of literature, including retrospective studies and subset analyses from three prospective studies, indicates that pretreatment CD19 expression is not strongly correlated with the efficacy of various CD19-directed therapies (48). Retrospective studies evaluating CD19 expression levels, as assessed by quantitative immunofluorescence analysis and IHC, have broadly shown that the ORR of CD19-directed therapies is independent of baseline CD19 expression, irrespective of the CD19-directed treatment modality, including CAR-T. In the phase II JULIET and ZUMA-2 trials, CD19 expression was assessed using quantitative immunofluorescence and IHC, respectively; these studies showed equivalent ORRs in patients with unequivocal CD19 expression and those with pretreatment low/negative CD19 expression (29, 49). Similarly, a phase II LOTIS-2 study evaluated CD19 expression (method not described) and reported that responses to loncastuximab tesirine were observed at all levels of CD19 expression in patients (50).

IHC and immunofluorescence are less sensitive and quantitative than conventional flow cytometry analysis, which offers a larger linear dynamic range for the determination of CD19 antigen expression than IHC (51, 52). A small single-center study found that a quantitative analysis of CD19 expression using flow cytometry did not identify a correlation between CD19 expression prior to CD19 CAR-T and outcomes (48). More studies are needed to verify these findings. However, flow cytometry is limited to liquid biopsies and is not routinely used for solid tissue diagnostics, such as lymphoma specimens, as the majority of diagnoses are performed on formalin-fixed, paraffin-embedded blocks. Additionally, these methodologies are difficult to clinically validate and are not routinely available in clinical laboratories.

In the context of CAR-T–cell responses, a limited number of preclinical studies have shown that the activity of CAR-T cells is dependent on target antigen density. CD19 antigen density can be specifically measured using calibration beads for flow cytometry with known fluorescent molecules per bead, as well as the number of fluorescent molecules conjugated to each antibody. For instance, CD19–CD28ζ CAR-T–cell activity was found to be highly dependent on CD19 antigen density (53). Of note, in preclinical models, CD19–CD28ζ CARs were more efficient at targeting CD19-low tumor cells than CD19-4-1BBζ CARs (53).

Treatment impact on expression levels

Changes between pretreatment and posttreatment CD19 expression in tumor biopsies may offer further insight into the dynamics of CD19 expression following CD19-directed therapy. A small retrospective exploratory analysis of the phase II L-MIND study used IHC, whole exome DNA, and exome RNA sequencing to quantify pretreatment and posttreatment CD19 expression and mutations. Five of six patients treated with tafasitamab retained comparable CD19 expression at both time points, with no evidence of CD19 genomic alterations. Of the six patients, three experienced an objective response (54). Similar data were reported for tafasitamab in an analysis of patients with R/R chronic lymphocytic leukemia, in which CD19 expression was maintained pretreatment and posttreatment (55, 56). A small single-center study examining patients who received axi-cel found that 10 of the 16 patients with LBCL who developed progressive disease after treatment with axi-cel had no or low CD19 expression (48). However, this is not the case for all CD19-targeted therapies in all relevant diseases; for example, DLBCL that relapses after CAR-T can be CD19 negative (48, 5760).

A case series that indicates the time point at which a tumor biopsy is collected following CD19-directed therapy may confound accurate assessment of posttreatment CD19 expression through temporary antigen masking (61). Two patients with CD19+ non-Hodgkin lymphoma (NHL) treated with tafasitamab underwent two posttreatment tumor biopsies within 1 and 3 to 4 weeks following the final tafasitamab dose. Interestingly, CD19 expression by IHC and flow cytometry was negative in the immediate posttreatment biopsy but was detected on the subsequent biopsy obtained at a later time point. These patients proceeded with CD19-directed CAR-T in later therapy lines. In this case, the half-life of tafasitamab may have resulted in persistent binding to CD19 at the time of the 1-week posttreatment biopsy, obscuring CD19 detection. To mitigate potential CD19 antigen masking, a suitable washout period may prove valuable for determining an accurate CD19 expression status following progression on CD19-directed therapy.

Although the data are intriguing, these studies included heterogeneous populations, used different mechanisms of assessing CD19 expression levels, lacked a uniform definition of CD19 positivity, and assessed posttreatment CD19 levels at varying time points. Further studies are required to determine the most clinically relevant mechanism and time points for assessing CD19 expression, particularly following CAR-T, because this area has not been adequately explored.

Although approximately 40% of patients experience a durable response to CAR-T, the remaining patients require subsequent lymphoma-directed treatment. Outcomes in the CAR-T refractory setting are poor, with mPFS and mOS of 2.8 and 5 to 9 months, respectively (62, 63). Well-established guidelines for optimal salvage treatment strategies are necessary.

Antigen escape

Loss of CD19 expression after CD19-directed CAR-T occurs in approximately 30% of patients with DLBCL (48, 5759) and is an important cause of therapeutic failure that would affect subsequent CD19-directed therapy. In a small study, 9 of 15 patients whose disease progressed following CAR-T had decreased surface CD19 levels, as measured using quantitative flow cytometry, and five of those nine patients had no detectable surface CD19 expression (<3,000 molecules per cell; ref. 48). The current understanding of CD19-directed antigen escape mechanisms is largely derived from studies in patients with B-cell acute lymphoblastic leukemia undergoing CAR-T and includes CD19 mutation(s) and splice variants and defective trafficking of CD19 to the cell surface membrane. CD19 mutations or splice variations (e.g., in exon 2 of the CD19 gene) can lead to a conformational change and loss of the CAR-T–cell CD19 epitope (64). Epitope mapping of the CD19 extracellular domain confirmed that the antigen-recognition domains of CAR-T cells (based on the FMC63 antibody) and tafasitamab (based on the 4G7 antibody) have partially overlapping conformationally sensitive epitopes dependent on spatially adjacent loops coded by exons 3 and 4, and splice variations in exon 2 could lead to epitope loss for CAR-T and tafasitamab (65, 66). Defective trafficking of CD19 to the cell surface as an antigen escape mechanism was reported in a patient with a CD19-negative acute lymphoblastic leukemia relapse after treatment with blinatumomab. Molecular evaluation showed the absence of CD81, a protein that regulates CD19 protein maturation and cell surface trafficking (67).

CD19-independent mechanisms of CAR-T failure

Using various genetic techniques, several groups have demonstrated that tumor features before CAR-T can help predict therapy outcomes, irrespective of pretreatment CD19 expression or CD19 genetic alterations. It has been reported that tumors with complex genomics have inferior responses to CD19 CAR-T (68, 69). Additionally, specific mutational patterns, such as those in RHOA and TMEM30A, were shown to affect outcomes in patients with these tumors (69, 70). Exploring how these features affect CD19-directed antibody therapies requires further investigation.

In addition to antigen escape, other mechanisms such as immunosuppression by the tumor microenvironment (TME), tumor metabolic volume at treatment, and intrinsic CAR-T–cell dysfunction have been shown to be involved in CAR-T failure in LBCL (71). Similar to patient endogenous T cells, CAR-T cells can also exhibit exhaustion, as measured by increased coinhibitory receptors and metabolic dysfunction (72). A CD8 T-cell exhaustion signature, as measured by single-cell RNA sequencing on CAR-T cells 7 days postadministration, was associated with a poor outcome in a study of 24 patients (73). Additionally, a CD8 memory phenotype in the infused CAR-T product was associated with superior outcomes. T-cell exhaustion has been proposed to account for the modest effect (27% ORR) of pembrolizumab, a programmed death-1 inhibitor, in a small prospective study of patients with DLBCL in whom CAR-T had failed (74). Additionally, the TME contains suppressive immune cell subsets such as regulatory T cells and myeloid-derived suppressor cells that can inhibit infiltrating CAR-T–cell function (75, 76) through nutrient and metabolite modulation as well as growth factor sequestration. T-cell metabolism and attempts to manufacture CAR-T cells to function in this inhospitable environment are beyond the scope of this publication but have been examined in several other reviews (7781).

CD19-directed therapy post–CAR-T relapse

CAR-T–cell persistence, T-cell exhaustion, and an immunosuppressive TME are likely prominent drivers of CAR-T failure, and limited data indicate that antigen escape is a less common etiology (82). Thus, subsequent CD19-directed therapy with an alternative mechanism of action is a rational approach for patients whose disease relapses after CAR-T. Data on the clinical utility of CD19-directed therapy for R/R DLBCL in the post–CAR-T setting are limited and primarily derived from real-world retrospective studies. A retrospective analysis of patients with CAR-T–refractory B-cell NHL treated with tafasitamab plus lenalidomide after CAR-T progression reported ORRs of 33% and 17% in the first- (n = 6) and second-line (n = 6) post–CAR-T treatment settings, respectively (63). A multicenter observational study reported an ORR of 17% and 10% for tafasitamab plus lenalidomide and loncastuximab tesirine, respectively (60), in patients with post–CAR-T R/R disease. In the LOTIS-2 study, an ORR of 42.9% was reported in 14 patients with post–CAR-T progression who subsequently received loncastuximab tesirine (27). Although these data indicate a potential role for CD19-directed therapy in the post–CAR-T setting, prospective studies with larger cohorts are necessary to guide clinicians.

The CAR-T–cell manufacture time is a key consideration for CAR-T administration. Bridging therapy is often necessary (8) and commonly consists of chemotherapy, radiation, steroids, or novel agents. Currently, data on CD19-directed antibodies, or ADCs, as a bridging therapy is lacking.

Limited data evaluating the efficacy of CAR-T in patients previously exposed to CD19-directed antibodies or ADCs are available. Preclinical evidence from a mouse xenograft model of mantle cell lymphoma indicates that tafasitamab has no detrimental effect on the efficacy of subsequent CAR-T–cell treatment (83). Using a CAR-T–cell construct similar to tisa-cel, mice treated sequentially with tafasitamab followed by CAR-T showed improved tumor control, later and stronger T-cell expansion, and a higher probability of survival than mice treated with saline followed by CAR-T. A second preclinical study indicated that pretreatment with tafasitamab before CD19 CAR-T improved efficacy while attenuating cytokine release syndrome (84). In a case study from the L-MIND trial, a patient whose disease relapsed following treatment with tafasitamab plus lenalidomide subsequently received axi-cel and experienced a CR (85). A retrospective analysis combining patients with R/R DLBCL from the phase I and II studies of loncastuximab tesirine who subsequently proceeded to CD19-directed CAR-T after disease relapse (n = 14) reported an ORR of 50% [CR = 6, partial response (PR) = 1] at the 3-month assessment (86). The median time between loncastuximab tesirine treatment and CAR-T was 120 days. Ten patients were evaluated for CD19 expression using IHC, and all showed retention of CD19 expression following loncastuximab tesirine treatment. The four patients with unknown posttreatment CD19 status following loncastuximab tesirine treatment had a CR with CAR-T, indicating that they also retained CD19 expression at progression. The phase III TRANSCEND trial investigating liso-cel in patients with R/R DLBCL included 12 patients with prior CD19-directed therapy [anti–CD19 monoclonal antibodies (n = 3), ADCs (n = 8), and bispecific T-cell engagers (n = 1)] and biopsy-confirmed persistent CD19 expression (87). A post hoc analysis of this subset showed that 11 of 12 patients (92%) achieved an objective response, five of whom had a response duration of ≥9 months and four were ongoing at the time of data cutoff.

The half-lives for tafasitamab and loncastuximab tesirine are ∼16 (88) and ∼21 (89) days, respectively, indicating that only 2% and 5% of antibodies will remain 90 days posttreatment, respectively. In addition to residual binding of CD19 from immediate pre–CAR-T, the recycling and production of new CD19 antigens following cessation of CD19-directed therapy may influence targetable antigen levels. However, these dynamics are not well characterized. There are no data to indicate a washout period between CD19-directed therapy and subsequent CAR-T. Further clinical studies are required to evaluate the optimal timing of sequential CD19-directed therapies.

Although much focus is placed on disease-related characteristics when selecting lymphoma-directed therapy, patient-related factors have an important role and include performance status, comorbidities, prior treatment, access to cellular therapy centers, psychosocial factors, and patient preference. Although only 50% of patients with R/R DLBCL are eligible for ASCT, the proportion of patients eligible for CAR-T remains undefined because of the lack of consensus regarding patient eligibility criteria (90). Using the ZUMA-1 inclusion/exclusion criteria, a single-center retrospective analysis performed in Sweden found that 49/60 (82%) patients with R/R DLBCL would have been eligible for CAR-T (91). This proportion stands in contrast to real-world findings in a multicenter retrospective analysis of axi-cel used as SOC for R/R LBCL, in which only 57% of 298 patients who received treatment met the ZUMA-1 criteria (42). In this study, 20% of patients did not meet the ZUMA-1 criteria because of an Eastern Cooperative Oncology Group performance score (ECOG PS) of >2, and the rest likely had medical comorbidities or were aged >65 years, which would have excluded them, although these data are not reported. However, despite not meeting the ZUMA-1 criteria, patients receiving axi-cel still experienced comparable safety and efficacy. Another real-world retrospective study found that only 49% of 1,500 patients met the ZUMA-1 criteria (92). In this study, the primary reason for patients not meeting the ZUMA-1 criteria was age >65 years in 38% of patients. An additional study investigating 129 patients whose disease progressed after frontline therapy estimated that 65% of patients would likely be eligible for second-line CAR-T if the trial inclusion criteria were expanded to an ECOG PS ≤2 (93). Considering patients aged >65 years and permitting an ECOG PS ≤2, ∼70% to 75% of patients in the R/R DLBCL setting have an ECOG PS of 0 to 1; thus, it is likely that a majority of patients with R/R DLBCL are eligible for CAR-T (94, 95).

ECOG PS is a key prognostic indicator in R/R DLBCL, and notably, patients with an ECOG PS of ≥2 were excluded from the three CAR-T registrational studies. The median age of patients in ZUMA-1, JULIET, and TRANSCEND was <60 years, whereas real-world data show that CAR-T is administered to and has comparable efficacy in patients aged 65 to 74 years, with poorer outcomes in those aged ≥75 years (96, 97). Retrospective real-world experience has also reported poorer outcomes in patients with an ECOG PS of ≥2 (42, 98, 99). For this patient population, CAR-T may not be the optimal CD19-directed therapy. Earlier treatment lines may influence the number and quality of autologous CAR-T cells. In particular, growing evidence suggests prior bendamustine-containing regimens negatively impact T-cell numbers and composition at apheresis and CAR-T–cell expansion, potentially reducing the efficacy of CAR-T–cell treatment (43, 100).

Unequal access to health care and logistical challenges further limit the use of CAR-T. At present, CAR-T is administered at a limited number of academic centers, often necessitating prolonged commutes, hospitalization, and caregiver support for extended periods, which are features that may limit this therapy for a subset of patients (101). Currently, 96.8% of CAR-T cases occur in urban hospitals, and although 19% of patients live in rural areas, only 14.7% of patients receiving CAR-T are from rural areas (102). This disparity is likely because of the need for close access to a cellular therapy center. Although over a third of patients live >2 hours away from a CAR-T center, patients living >60 miles from a CAR-T center were less likely to undergo therapy than those living <60 miles (103). Geographical concerns are also at play; in the United States, patients in the South were less likely to have access to CAR-T than those in the Northeast (OR = 0.284; ref. 104). Income level and insurance status also affect access to CAR-T. Patients with higher income levels are more likely to receive CAR-T, with one study showing that only 7.3% of patients who receive CAR-T come from neighborhoods with a median income of less than $40,000. Similarly, patients receiving CAR-T are less likely to have Medicare or be uninsured, which limits access to treatment because of the high cost of therapy (103).

BsAb therapies are also gaining approval for R/R DLBCL. Although these products are “off-the-shelf” and do not require manufacturing for individual patients, there are limitations to their widespread adoption in clinical practice. Similar to CAR-T, these treatments require hospitalization for administration or step-up dosing, which limits their use in certain settings. However, an ongoing clinical trial is challenging the role of inpatient step-up dosing for epcoritimab (NCT05451810). Although long-term data from CAR-T–cell trials such as ZUMA-1 have shown a flattening in progression-free survival/overall survival curves, indicating that ∼40% of patients may be cured, long-term data on BsAbs are lacking. For patients with either disease- or patient-related factors that are less suitable for the logistical and toxicity challenges of CD19 CAR-T or BsAbs, more readily available “off-the-shelf” CD19-directed therapies administered in the outpatient setting, such as tafasitamab plus lenalidomide and loncastuximab tesirine, represent an alternative approach (105). These regimens often have fewer burdensome side effects and have promising clinical efficacy.

As approved indications for CD19-directed therapies expand, clinicians will be increasingly faced with the dilemma of optimal use. Several factors influence optimal treatment sequencing. For patients with a suitable degree of fitness and access to limited centers that provide immune effector cell therapy, CAR-T will likely be the initial CD19-directed therapy. For more frail patients, those with rapidly progressive disease, or those without access to tertiary referral centers, monoclonal antibodies, or ADCs, may be the initial CD19-directed therapy.

Defining clinically relevant thresholds and sensitive CD19 expression level quantification methodologies will be paramount for optimizing sequencing. Given the limitations of routinely available CD19 assays and the potential for antigen masking or reductions in CD19 expression following CD19-directed therapy, a suitable washout period may be indicated to obtain an accurate posttreatment disease phenotype. However, no guidelines are available to define the appropriate timeline for a treatment washout, and the urgency of treatment due to patients’ disease characteristics may be the primary driver of subsequent treatment selection.

Genomic classification of DLBCL has shown utility in predicting responses to R-CHOP chemotherapy and may have utility in predicting responses to other targeted therapies (106109). However, these genetic subgroups have not shown utility in predicting outcomes with CD19-directed therapies, and novel predictors for CD19-directed therapies are needed.

In patients whose disease progresses after CAR-T, responses to currently available treatments are suboptimal, and enrollment in a clinical trial is recommended. As approximately 30% of patients with DLBCL experience relapse with CD19-negative disease following CAR-T (5759), reassessment of CD19 expression should be strongly considered to inform subsequent treatment strategies. Although retrospective data indicate that CD19-directed therapy may offer clinical benefit in a subset of patients following progression after CAR-T with retained CD19 expression (26, 60, 63), prospective studies are needed.

Limited clinical data indicate that CD19-directed therapies do not preclude the future successful application of CAR-T (54, 8587). There are insufficient data to recommend the use of CD19-directed therapy as a bridging therapy for CAR-T.

In summary, there are limited clinical data to guide definitive decision-making for sequencing CD19-directed therapies in patients with R/R DLBCL. A sufficiently powered and appropriately designed clinical or real-world study, with the involvement of both clinical institutions and pharmaceutical companies, would substantially aid in the determination of the effectiveness of subsequent CAR-T in patients with R/R DLBCL who received prior CD19-directed therapy. For ongoing and future clinical studies, the collection of sequential CD19 expression data for correlation with clinical responses and the likelihood of responses to sequential CD19-directed therapy are urgently required to inform further decision-making to best benefit patients with DLBCL.

R. Birhiray reports personal fees from Incyte, MorphoSys, GSK, Pharmacyclics, Janssen, AbbVie, BeiGene, Lilly Oncology, Exelixis, Puma, and ADC Therapeutics outside the submitted work. A. Merchant reports personal fees from AbbVie, Novartis, and Oncovalent outside the submitted work. No disclosures were reported by the other authors.

Medical writing and editorial support for the development of this article were provided by Stuart Langley of Syneos Health, UK, and funded by MorphoSys AG, in accordance with Good Publication Practice. MorphoSys AG conducted a courtesy review for the scientific accuracy of the manuscript before submission but had no role in the development of the content, selection of the journal for submission, or the decision to submit the manuscript for publication.

1.
Coiffier
B
,
Thieblemont
C
,
Van Den Neste
E
,
Lepeu
G
,
Plantier
I
,
Castaigne
S
, et al
.
Long-term outcome of patients in the LNH-98.5 trial, the first randomized study comparing rituximab-CHOP to standard CHOP chemotherapy in DLBCL patients: a study by the Groupe d'Etudes des Lymphomes de l'Adulte
.
Blood
2010
;
116
:
2040
5
.
2.
Food & Drug Administration
.
Rituximab. Prescribing information
[
Internet
].
2021
[cited 2022 Aug 1]. Available from:
www.accessdata.fda.gov/drugsatfda_docs/label/2021/103705s5467lbl.pdf.
3.
Food & Drug Administration
.
Cyclophosphamide. Prescribing information
[
Internet
].
2021
[cited 2022 Aug 1]. Available from:
www.accessdata.fda.gov/drugsatfda_docs/label/2021/212501s002lbl.pdf.
4.
Food & Drug Administration
.
Doxorubicin. Prescribing information
[
Internet
].
2019
[cited 2022 Aug 1]. Available from:
www.accessdata.fda.gov/drugsatfda_docs/label/2020/050467s078,050629s030lbl.pdf.
5.
Food & Drug Administration
.
Vincristine. Prescribing information
[
Internet
].
2013
[cited 2022 Aug 1]. Available from:
www.accessdata.fda.gov/drugsatfda_docs/label/2014/071484s042lbl.pdf.
6.
Food & Drug Administration
.
Prednisolone. Prescribing information
[
Internet
].
2010
[cited 2022 Aug 1]. Available from:
www.accessdata.fda.gov/drugsatfda_docs/label/2010/021959s004lbl.pdf.
7.
Sarkozy
C
,
Sehn
LH
.
Management of relapsed/refractory DLBCL
.
Best Pract Res Clin Haematol
2018
;
31
:
209
16
.
8.
NCCN Clinical Practice Guidelines in Oncology
.
B-Cell Lymphomas V.2.2023
.
[cited 2023 Feb 24]
.
9.
Food & Drug Administration
.
COLUMVI. Prescribing information
[
Internet
].
2023
[cited 2023 Jun 21]. Available from:
https://www.gene.com/download/pdf/columvi_prescribing.pdf.
10.
Food & Drug Administration
.
EPKINLY. Prescribing information
[
Internet
].
2023
[cited 2023 Feb 24]. Available from:
https://www.accessdata.fda.gov/drugsatfda_docs/label/2023/761324s000lbl.pdf.
11.
Ayyappan
S
,
Kim
WS
,
Kim
TM
,
Walewski
J
,
Cho
S-G
,
Jarque
I
, et al
.
Final analysis of the phase 2 ELM-2 study: odronextamab in patients with relapsed/refractory (R/R) diffuse large B-cell lymphoma (DLBCL)
.
Blood
2023
;
142
:
436
.
12.
Russler-Germain
DA
,
Ghobadi
A
.
T-cell redirecting therapies for B-cell non-Hodgkin lymphoma: recent progress and future directions
.
Front Oncol
2023
;
13
:
1168622
.
13.
Duell
J
,
Abrisqueta
P
,
Andre
M
,
Gaidano
G
,
Gonzales-Barca
E
,
Jurczak
W
, 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
2023
;
109
:
553
66
.
14.
Westin
JR
,
Oluwole
OO
,
Kersten
MJ
,
Miklos
DB
,
Perales
M-A
,
Ghobadi
A
, et al
.
Survival with axicabtagene ciloleucel in large B-cell lymphoma
.
N Engl J Med
2023
;
389
:
148
57
.
15.
Schuster
SJ
,
Tam
CS
,
Borchmann
P
,
Worel
N
,
McGuirk
JP
,
Holte
H
, 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
15
.
16.
Wang
K
,
Wei
G
,
Liu
D
.
CD19: a biomarker for B cell development, lymphoma diagnosis and therapy
.
Exp Hematol Oncol
2012
;
1
:
36
.
17.
Pavlasova
G
,
Mraz
M
.
The regulation and function of CD20: an “enigma” of B-cell biology and targeted therapy
.
Haematologica
2020
;
105
:
1494
506
.
18.
Horna
P
,
Nowakowski
G
,
Endell
J
,
Boxhammer
R
.
Comparative assessment of surface CD19 and CD20 expression on B-cell lymphomas from clinical biopsies: implications for targeted therapies
.
Blood
2019
;
134
:
5345
.
19.
Marshalek
JP
,
Dragan
M
,
Tomassetti
S
,
LaBarbera
K
,
Amaya
A
.
CD20 and CD19 expression loss in relapsed or refractory B-cell non-Hodgkin lymphoma: a retrospective cohort
.
J Clin Oncol
2022
;
40
:
e19537
.
20.
Vitolo
U
,
Nowakowski
GS
,
Burke
JM
,
Fox
CP
,
Trneny
M
,
Chiappella
A
, et al
.
ABCL-021: front-mind: a phase III, randomized, double-blind, placebo-controlled study comparing efficacy and safety of tafasitamab + lenalidomide + R-CHOP vs R-CHOP alone for newly-diagnosed high-intermediate and high-risk diffuse large B-cell lymphoma
.
Clin Lymphoma Myeloma Leuk
2021
;
21
:
S376
-
7
.
21.
Food & Drug Administration
.
Tafasitamab. Prescribing information
[
Internet
].
2021
[cited 2022 Aug 1]. Available from:
www.accessdata.fda.gov/drugsatfda_docs/label/2021/761163s001lbl.pdf.
22.
Food & Drug Administration
.
Lenalidomide
. Prescribing information [
Internet
].
2013
[cited 2022 Aug 1]. Available from:
www.accessdata.fda.gov/drugsatfda_docs/label/2013/021880s034lbl.pdf.
23.
Salles
G
,
Duell
J
,
González Barca
E
,
Tournilhac
O
,
Jurczak
W
,
Liberati
AM
, et al
.
Tafasitamab plus lenalidomide in relapsed or refractory diffuse large B-cell lymphoma (L-MIND): a multicentre, prospective, single-arm, phase 2 study
.
Lancet Oncol
2020
;
21
:
978
88
.
24.
Hartley
JA
,
Flynn
MJ
,
Bingham
JP
,
Corbett
S
,
Reinert
H
,
Tiberghien
A
, et al
.
Pre-clinical pharmacology and mechanism of action of SG3199, the pyrrolobenzodiazepine (PBD) dimer warhead component of antibody-drug conjugate (ADC) payload tesirine
.
Sci Rep
2018
;
8
:
10479
.
25.
Food & Drug Administration
.
Loncastuximab tesirine. Prescribing information
[
Internet
].
2021
[cited 2022 Aug 1]. Available from:
www.accessdata.fda.gov/drugsatfda_docs/label/2021/761196s000lbl.pdf.
26.
Caimi
PF
,
Ai
W
,
Alderuccio
JP
,
Ardeshna
KM
,
Hamadani
M
,
Hess
B
, et al
.
Loncastuximab tesirine in relapsed or refractory diffuse large B-cell lymphoma (LOTIS-2): a multicentre, open-label, single-arm, phase 2 trial
.
Lancet Oncol
2021
;
22
:
790
800
.
27.
Caimi
PF
,
Ai
WZ
,
Alderuccio
JP
,
Ardeshna
KM
,
Hamadani
M
,
Hess
B
, et al
.
Loncastuximab tesirine in relapsed/refractory diffuse large B-cell lymphoma: long-term efficacy and safety from the phase II LOTIS-2 study
.
Haematologica
2024
;
109
:
1184
93
.
28.
Locke
FL
,
Neelapu
SS
,
Bartlett
NL
,
Siddiqi
T
,
Chavez
JC
,
Hosing
CM
, et al
.
Phase 1 results of ZUMA-1: a multicenter study of KTE-C19 anti-CD19 CAR T cell therapy in refractory aggressive lymphoma
.
Mol Ther
2017
;
25
:
285
95
.
29.
Schuster
SJ
,
Bishop
MR
,
Tam
CS
,
Waller
EK
,
Borchmann
P
,
McGuirk
JP
, et al
.
Tisagenlecleucel in adult relapsed or refractory diffuse large B-cell lymphoma
.
N Engl J Med
2019
;
380
:
45
56
.
30.
Abramson
JS
.
Anti-CD19 CAR T-cell therapy for B-cell non-Hodgkin lymphoma
.
Transfus Med Rev
2020
;
34
:
29
33
.
31.
Eshhar
Z
,
Waks
T
,
Gross
G
,
Schindler
DG
.
Specific activation and targeting of cytotoxic lymphocytes through chimeric single chains consisting of antibody-binding domains and the gamma or zeta subunits of the immunoglobulin and T-cell receptors
.
Proc Natl Acad Sci U S A
1993
;
90
:
720
4
.
32.
Tyagarajan
S
,
Spencer
T
,
Smith
J
.
Optimizing CAR-T cell manufacturing processes during pivotal clinical trials
.
Mol Ther Methods Clin Dev
2020
;
16
:
136
44
.
33.
Neelapu
SS
,
Locke
FL
,
Bartlett
NL
,
Lekakis
LJ
,
Miklos
DB
,
Jacobson
CA
, et al
.
Axicabtagene ciloleucel CAR T-cell therapy in refractory large B-cell lymphoma
.
N Engl J Med
2017
;
377
:
2531
44
.
34.
Neelapu
SS
,
Jacobson
CA
,
Ghobadi
A
,
Miklos
DB
,
Lekakis
LJ
,
Oluwole
OO
, 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
15
.
35.
Locke
FL
,
Miklos
DB
,
Jacobson
CA
,
Perales
M-A
,
Kersten
M-J
,
Oluwole
OO
, et al
.
Axicabtagene ciloleucel as second-line therapy for large B-cell lymphoma
.
N Engl J Med
2022
;
386
:
640
54
.
36.
Bishop
MR
,
Dickinson
M
,
Purtill
D
,
Barba
P
,
Santoro
A
,
Hamad
N
, et al
.
Second-line tisagenlecleucel or standard care in aggressive B-cell lymphoma
.
N Engl J Med
2022
;
386
:
629
39
.
37.
Abramson
JS
,
Palomba
ML
,
Gordon
LI
,
Lunning
MA
,
Wang
M
,
Arnason
J
, 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
52
.
38.
Abramson
JS
,
Palomba
ML
,
Gordon
LI
,
Lunning
MA
,
Wang
M
,
Arnason
JE
, 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
.
39.
Abramson
JS
,
Solomon
SR
,
Arnason
JE
,
Johnston
PB
,
Glass
B
,
Bachanova
V
, et al
.
Lisocabtagene maraleucel as second-line therapy for large B-cell lymphoma: primary analysis of phase 3 TRANSFORM study
.
Blood
2023
;
141
:
1675
84
.
40.
Vercellino
L
,
Di Blasi
R
,
Kanoun
S
,
Tessoulin
B
,
Rossi
C
,
D’Aveni-Piney
M
, et al
.
Predictive factors of early progression after CAR T-cell therapy in relapsed/refractory diffuse large B-cell lymphoma
.
Blood Adv
2020
;
4
:
5607
15
.
41.
Jacobson
CA
,
Hunter
BD
,
Redd
R
,
Rodig
SJ
,
Chen
P-H
,
Wright
K
, et al
.
Axicabtagene ciloleucel in the non-trial setting: outcomes and correlates of response, resistance, and toxicity
.
J Clin Oncol
2020
;
38
:
3095
106
.
42.
Nastoupil
LJ
,
Jain
MD
,
Feng
L
,
Spiegel
JY
,
Ghobadi
A
,
Lin
Y
, et al
.
Standard-of-care axicabtagene ciloleucel for relapsed or refractory large B-cell lymphoma: results from the US lymphoma CAR T consortium
.
J Clin Oncol
2020
;
38
:
3119
28
.
43.
Schuster
SJ
,
Tam
CS
,
Borchmann
P
,
Worel
N
,
McGuirk
JP
,
Holte
H
, 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
15
.
44.
Tilly
H
,
Gomes da Silva
M
,
Vitolo
U
,
Jack
A
,
Meignan
M
,
Lopez-Guillermo
A
, et al
.
Diffuse large B-cell lymphoma (DLBCL): ESMO clinical practice guidelines for diagnosis, treatment and follow-up
.
Ann Oncol
2015
;
26
:
v116
-
25
.
45.
D’Arena
G
,
Musto
P
,
Cascavilla
N
,
Dell’Olio
M
,
Di Renzo
N
,
Carotenuto
M
.
Quantitative flow cytometry for the differential diagnosis of leukemic B-cell chronic lymphoproliferative disorders
.
Am J Hematol
2000
;
64
:
275
81
.
46.
Riva
G
,
Nasillo
V
,
Ottomano
AM
,
Bergonzini
G
,
Paolini
A
,
Forghieri
F
, et al
.
Multiparametric flow cytometry for MRD monitoring in hematologic malignancies: clinical applications and new challenges
.
Cancers (Basel)
2021
;
13
:
4582
.
47.
Rimm
DL
.
What brown cannot do for you
.
Nat Biotechnol
2006
;
24
:
914
6
.
48.
Spiegel
JY
,
Patel
S
,
Muffly
L
,
Hossain
NM
,
Oak
J
,
Baird
JH
, et al
.
CAR T cells with dual targeting of CD19 and CD22 in adult patients with recurrent or refractory B cell malignancies: a phase 1 trial
.
Nat Med
2021
;
27
:
1419
31
.
49.
Wang
M
,
Munoz
J
,
Goy
A
,
Locke
FL
,
Jacobson
CA
,
Hill
BT
, et al
.
KTE-X19 CAR T-cell therapy in relapsed or refractory mantle-cell lymphoma
.
N Engl J Med
2020
;
382
:
1331
42
.
50.
Caimi
PF
,
Hamadani
M
,
Carlo-Stella
C
,
Nickaeen
M
,
Jordie
E
,
Utsey
K
, et al
.
CD19 expression by IHC alone is not a predictor of response to loncastuximab tesirine: results from the LOTIS-2 clinical trial and quantitative systems pharmacology modeling
.
Blood
2022
;
140
:
9548
50
.
51.
van Dam
LS
,
Oskam
JM
,
Kamerling
SWA
,
Arends
EJ
,
Bredewold
OW
,
Berkowska
MA
, et al
.
Highly sensitive flow cytometric detection of residual B-cells after rituximab in anti-neutrophil cytoplasmic antibodies-associated vasculitis patients
.
Front Immunol
2020
;
11
:
566732
.
52.
Zola
H
.
High-sensitivity immunofluorescence/flow cytometry: detection of cytokine receptors and other low-abundance membrane molecules
.
Curr Protoc Cytom
2004
;
Chapter 6
:
Unit 6.3
.
53.
Majzner
RG
,
Rietberg
SP
,
Sotillo
E
,
Dong
R
,
Vachharajani
VT
,
Labanieh
L
, et al
.
Tuning the antigen density requirement for CAR T-cell activity
.
Cancer Discov
2020
;
10
:
702
23
.
54.
Duell
J
,
Obr
A
,
Augustin
M
,
Endell
J
,
Liu
H
,
Geiger
S
, et al
.
CD19 expression is maintained in DLBCL patients after treatment with tafasitamab plus lenalidomide in the L-MIND study
.
Leuk Lymphoma
2022
;
63
:
468
72
.
55.
Woyach
JA
,
Awan
F
,
Flinn
IW
,
Berdeja
JG
,
Wiley
E
,
Mansoor
S
, et al
.
A phase 1 trial of the Fc-engineered CD19 antibody XmAb5574 (MOR00208) demonstrates safety and preliminary efficacy in relapsed CLL
.
Blood
2014
;
124
:
3553
60
.
56.
Boxhammer
R
,
Striebel
F
,
Baumgartner
R
,
Endell
J
.
Expression of CD19 antigen on chronic lymphocytic leukemia cells after tafasitamab (anti-CD19) treatment: phase I trial data
.
Blood
2019
;
134
:
5061
.
57.
Plaks
V
,
Rossi
JM
,
Chou
J
,
Wang
L
,
Poddar
S
,
Han
G
, et al
.
CD19 target evasion as a mechanism of relapse in large B-cell lymphoma treated with axicabtagene ciloleucel
.
Blood
2021
;
138
:
1081
5
.
58.
Spiegel
JY
,
Dahiya
S
,
Jain
MD
,
Tamaresis
J
,
Nastoupil
LJ
,
Jacobs
MT
, et al
.
Outcomes of patients with large B-cell lymphoma progressing after axicabtagene ciloleucel therapy
.
Blood
2021
;
137
:
1832
5
.
59.
Neelapu
SS
,
Rossi
JM
,
Jacobson
CA
,
Locke
FL
,
Miklos
DB
,
Reagan
PM
, et al
.
CD19-loss with preservation of other B cell lineage features in patients with large B cell lymphoma who relapsed post-axi-cel
.
Blood
2019
;
134
:
203
.
60.
Iqbal
M
,
Khurana
A
,
Chavez
J
,
Rosenthal
AC
,
Li
Z
,
Craver
E
, et al
.
Efficacy of CD19 directed therapies in large B-cell lymphoma (LBCL) relapsing after chimeric antigen receptor (CAR) T-cell therapy
.
Blood
2022
;
140
:
10396
8
.
61.
Fitzgerald
KN
,
Quesada
AE
,
von Keudell
G
,
Raj
S
,
Lewis
NE
,
Dogan
A
, et al
.
CD19 epitope masking by tafasitamab leads to delays in subsequent use of CD19 CAR T-cell therapy in two patients with aggressive mature B-cell lymphomas
.
Leuk Lymphoma
2022
;
63
:
751
4
.
62.
Di Blasi
R
,
Le Gouill
S
,
Bachy
E
,
Cartron
G
,
Beauvais
D
,
Le Bras
F
, et al
.
Outcomes of patients with aggressive B-cell lymphoma after failure of anti-CD19 CAR T-cell therapy: a DESCAR-T analysis
.
Blood
2022
;
140
:
2584
93
.
63.
Zurko
J
,
Nizamuddin
I
,
Epperla
N
,
David
K
,
Cohen
JB
,
Moyo
TK
, 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
69
.
64.
Sotillo
E
,
Barrett
DM
,
Black
KL
,
Bagashev
A
,
Oldridge
D
,
Wu
G
, et al
.
Convergence of acquired mutations and alternative splicing of CD19 enables resistance to CART-19 immunotherapy
.
Cancer Discov
2015
;
5
:
1282
95
.
65.
Klesmith
JR
,
Wu
L
,
Lobb
RR
,
Rennert
PD
,
Hackel
BJ
.
Fine epitope mapping of the CD19 extracellular domain promotes design
.
Biochemistry
2019
;
58
:
4869
81
.
66.
Fischer
J
,
Paret
C
,
El Malki
K
,
Alt
F
,
Wingerter
A
,
Neu
MA
, et al
.
CD19 isoforms enabling resistance to CART-19 immunotherapy are expressed in B-ALL patients at initial diagnosis
.
J Immunother
2017
;
40
:
187
95
.
67.
Braig
F
,
Brandt
A
,
Goebeler
M
,
Tony
HP
,
Kurze
AK
,
Nollau
P
, et al
.
Resistance to anti-CD19/CD3 BiTE in acute lymphoblastic leukemia may be mediated by disrupted CD19 membrane trafficking
.
Blood
2017
;
129
:
100
4
.
68.
Jain
MD
,
Ziccheddu
B
,
Coughlin
CA
,
Faramand
R
,
Griswold
AJ
,
Reid
KM
, et al
.
Whole-genome sequencing reveals complex genomic features underlying anti-CD19 CAR T-cell treatment failures in lymphoma
.
Blood
2022
;
140
:
491
503
.
69.
Cherng
H-JJ
,
Sun
R
,
Sugg
B
,
Irwin
R
,
Yang
H
,
Le
CC
, et al
.
Risk assessment with low-pass whole-genome sequencing of cell-free DNA before CD19 CAR T-cell therapy for large B-cell lymphoma
.
Blood
2022
;
140
:
504
15
.
70.
Sworder
BJ
,
Kurtz
DM
,
Alig
SK
,
Frank
MJ
,
Shukla
N
,
Garofalo
A
, et al
.
Determinants of resistance to engineered T cell therapies targeting CD19 in large B cell lymphomas
.
Cancer Cell
2023
;
41
:
210
225.e5
.
71.
Dean
EA
,
Mhaskar
RS
,
Lu
H
,
Mousa
MS
,
Krivenko
GS
,
Lazaryan
A
, et al
.
High metabolic tumor volume is associated with decreased efficacy of axicabtagene ciloleucel in large B-cell lymphoma
.
Blood Adv
2020
;
4
:
3268
76
.
72.
Colombo
AR
,
Hav
M
,
Singh
M
,
Xu
A
,
Gamboa
A
,
Lemos
T
, et al
.
Single-cell spatial analysis of tumor immune architecture in diffuse large B-cell lymphoma
.
Blood Adv
2022
;
6
:
4675
90
.
73.
Deng
Q
,
Han
G
,
Puebla-Osorio
N
,
Ma
MCJ
,
Strati
P
,
Chasen
B
, 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
87
.
74.
Chong
EA
,
Svoboda
J
,
Dwivedy Nasta
S
,
Landsburg
DJ
,
Winchell
N
,
Napier
E
, et al
.
Sequential anti-CD19 directed chimeric antigen receptor modified T-cell therapy (CART19) and PD-1 blockade with pembrolizumab in patients with relapsed or refractory B-cell non-Hodgkin lymphomas
.
Blood
2018
;
132
:
4198
.
75.
Baumann
T
,
Dunkel
A
,
Schmid
C
,
Schmitt
S
,
Hiltensperger
M
,
Lohr
K
, et al
.
Regulatory myeloid cells paralyze T cells through cell–cell transfer of the metabolite methylglyoxal
.
Nat Immunol
2020
;
21
:
555
66
.
76.
Yan
Z
,
Garg
SK
,
Kipnis
J
,
Banerjee
R
.
Extracellular redox modulation by regulatory T cells
.
Nat Chem Biol
2009
;
5
:
721
3
.
77.
Fox
CJ
,
Hammerman
PS
,
Thompson
CB
.
Fuel feeds function: energy metabolism and the T-cell response
.
Nat Rev Immunol
2005
;
5
:
844
52
.
78.
Pearce
EL
,
Pearce
EJ
.
Metabolic pathways in immune cell activation and quiescence
.
Immunity
2013
;
38
:
633
43
.
79.
Sabatino
M
,
Hu
J
,
Sommariva
M
,
Gautam
S
,
Fellowes
V
,
Hocker
JD
, et al
.
Generation of clinical-grade CD19-specific CAR-modified CD8+ memory stem cells for the treatment of human B-cell malignancies
.
Blood
2016
;
128
:
519
28
.
80.
Quail
DF
,
Joyce
JA
.
Microenvironmental regulation of tumor progression and metastasis
.
Nat Med
2013
;
19
:
1423
37
.
81.
Klebanoff
CA
,
Gattinoni
L
,
Restifo
NP
.
CD8+ T-cell memory in tumor immunology and immunotherapy
.
Immunol Rev
2006
;
211
:
214
24
.
82.
Cheng
J
,
Zhao
L
,
Zhang
Y
,
Qin
Y
,
Guan
Y
,
Zhang
T
, et al
.
Understanding the mechanisms of resistance to CAR T-cell therapy in malignancies
.
Front Oncol
2019
;
9
:
1237
.
83.
Sakemura
R
,
Horvei
P
,
Manriquez-Roman
C
,
Ogbodo
EJ
,
Tapper
EE
,
Siegler
EL
, 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
.
84.
Sakemura
RL
,
Manriquez-Roman
C
,
Horvei
P
,
Cox
MJ
,
Huynh
T
,
Girsch
JH
, 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
4
.
85.
Oliai
C
,
de Vos
S
.
Case report: sustained remission achieved from anti-CD19 CAR T cell therapy despite prior treatment with anti-CD19 antibody tafasitamab (MOR208) in a patient with relapsed and refractory diffuse large B-cell lymphoma
.
Blood
2019
;
134
:
5360
.
86.
Thapa
B
,
Caimi
PF
,
Ardeshna
KM
,
Solh
M
,
Carlo-Stella
C
,
Kahl
BS
, et al
.
CD19 antibody-drug conjugate therapy in DLBCL does not preclude subsequent responses to CD19-directed CAR T-cell therapy
.
Blood Adv
2020
;
4
:
3850
2
.
87.
Solomon
SR
,
Mehta
A
,
Abramson
JS
,
Siddiqi
T
,
Lunning
M
,
Maloney
DG
, et al
.
Experience of prior anti-CD19 therapy in patients (Pts) with relapsed or refractory (R/R) large B-cell non-hodgkin lymphoma (B-NHL) receiving lisocabtagene maraleucel (liso-cel), an investigational anti-CD19 chimeric antigen receptor (CAR) T cell product
.
Transpl Cell Ther
2021
;
27
:
S207
-
8
.
88.
Jurczak
W
,
Zinzani
P
,
Gaidano
G
,
Goy
A
,
Provencio
M
,
Nagy
Z
, et al
.
Phase IIa study of single-agent MOR208 in patients with relapsed or refractory B-cell non-hodgkin's lymphoma
.
Blood
2015
;
126
:
1528
.
89.
Food & Drug Administration
.
ZYNLONTA (loncastuximab tesirine-lpyl). Prescribing information
. [
Internet
].
2021
[cited 2022 Apr 19]. Available from:
https://www.accessdata.fda.gov/drugsatfda_docs/label/2021/761196s000lbl.pdf.
90.
Gisselbrecht
C
,
Glass
B
,
Mounier
N
,
Gill
DS
,
Linch
DC
,
Trneny
M
, et al
.
Salvage regimens with autologous transplantation for relapsed large B-cell lymphoma in the rituximab era
.
J Clin Oncol
2010
;
28
:
4184
90
.
91.
Bailey
N
,
Pagel
JM
,
Bensinger
W
,
Hegerova
L
,
Mawad
R
,
Milder
M
, et al
.
A single center retrospective study of real world relapsed/refractory DLBCL patients eligible for CAR T-cell therapy: patient characteristics and outcomes
.
Blood
2018
;
132
:
1715
.
92.
Locke
FL
,
Jacobson
C
,
Ma
L
,
Dong
H
,
Hu
Z-H
,
Siddiqi
T
, et al
.
Real-world outcomes of axicabtagene ciloleucel (axi-cel) for the treatment of large B-cell lymphoma (LBCL): impact of age and specific organ dysfunction
.
Blood
2021
;
138
:
530
.
93.
Puckrin
R
,
Stewart
DA
,
Shafey
M
.
Real-world eligibility for second-line chimeric antigen receptor T cell therapy in large B cell lymphoma: a population-based analysis
.
Transpl Cell Ther
2022
;
28
:
218.e1
-
e4
.
94.
Harrysson
S
,
Eloranta
S
,
Ekberg
S
,
Enblad
G
,
Jerkeman
M
,
Wahlin
BE
, et al
.
Incidence of relapsed/refractory diffuse large B-cell lymphoma (DLBCL) including CNS relapse in a population-based cohort of 4243 patients in Sweden
.
Blood Cancer J
2021
;
11
:
9
.
95.
Crump
M
,
Neelapu
SS
,
Farooq
U
,
Van Den Neste
E
,
Kuruvilla
J
,
Westin
J
, et al
.
Outcomes in refractory diffuse large B-cell lymphoma: results from the international SCHOLAR-1 study
.
Blood
2017
;
130
:
1800
8
.
96.
Sano
D
,
Lekakis
L
,
Feng
L
,
Nastoupil
LJ
,
Jain
MD
,
Spiegel
JY
, et al
.
Safety and efficacy of axicabtagene ciloleucel (axi-cel) in older patients: results from the US lymphoma CAR-T consortium
.
Hematol Oncol
2019
;
37
:
304
5
.
97.
Chihara
D
,
Liao
L
,
Tkacz
J
,
Franco
A
,
Lewing
B
,
Kilgore
KM
, et al
.
Real-world experience of CAR T-cell therapy in older patients with relapsed/refractory diffuse large B-cell lymphoma
.
Blood
2023
;
142
:
1047
55
.
98.
Bastos-Oreiro
M
,
Gutiérrez
A
,
Reguera
JL
,
Iacoboni
G
,
López-Corral
L
,
Terol
MJ
, et al
.
Best treatment option for patients with refractory aggressive B-cell lymphoma in the CAR-T cell era: real-world evidence from GELTAMO/GETH Spanish groups
.
Front Immunol
2022
;
13
:
855730
.
99.
Jacobson
CA
,
Locke
FL
,
Ma
L
,
Asubonteng
J
,
Hu
Z-H
,
Siddiqi
T
, et al
.
Real-world evidence of axicabtagene ciloleucel for the treatment of large B-cell lymphoma in the United States
.
Transpl Cell Ther
2022
;
28
:
581.e1
-
e8
.
100.
Iacoboni
G
,
Lopez
AAM
,
Jalowiec
KA
,
Kwon
M
,
Rejeski
K
,
Garcés
VN
, et al
.
Recent bendamustine treatment before apheresis has a negative impact on outcomes in patients with large B-cell lymphoma receiving chimeric antigen receptor T-cell therapy
.
Blood
2022
;
140
:
1592
4
.
101.
Myers
GD
,
Verneris
MR
,
Goy
A
,
Maziarz
RT
.
Perspectives on outpatient administration of CAR-T cell therapy in aggressive B-cell lymphoma and acute lymphoblastic leukemia
.
J Immunother Cancer
2021
;
9
:
e002056
.
102.
Emole
J
,
Lawal
O
,
Lupak
O
,
Dias
A
,
Shune
L
,
Yusuf
K
.
Demographic differences among patients treated with chimeric antigen receptor T-cell therapy in the United States
.
Cancer Med
2022
;
11
:
4440
8
.
103.
Ahmed
N
,
Shahzad
M
,
Shippey
E
,
Bansal
R
,
Mushtaq
MU
,
Mahmoudjafari
Z
, et al
.
Socioeconomic and racial disparity in chimeric antigen receptor T cell therapy access
.
Transpl Cell Ther
2022
;
28
:
358
64
.
104.
Saunders
A
,
Inguva
S
,
Keating
SJ
,
Chirikov
V
.
Examination of disparities in access to chimeric antigen receptor (CAR) T cell therapies in Medicare patients (Pts) with relapsed or refractory (R/R) diffuse large B-cell lymphoma (DLBCL) who had failed at least 2 prior lines of therapy (LOT)
.
Blood
2022
;
140
:
10921
3
.
105.
Perrone
S
,
Lopedote
P
,
Levis
M
,
Di Rocco
A
,
Smith
SD
.
Management of relapsed or refractory large B-cell lymphoma in patients ineligible for CAR-T cell therapy
.
Expert Rev Hematol
2022
;
15
:
215
32
.
106.
Wilson
WH
,
Wright
GW
,
Huang
DW
,
Hodkinson
B
,
Balasubramanian
S
,
Fan
Y
, et al
.
Effect of ibrutinib with R-CHOP chemotherapy in genetic subtypes of DLBCL
.
Cancer Cell
2021
;
39
:
1643
53.e3
.
107.
Ren
W
,
Wan
H
,
Own
SA
,
Berglund
M
,
Wang
X
,
Yang
M
, et al
.
Genetic and transcriptomic analyses of diffuse large B-cell lymphoma patients with poor outcomes within two years of diagnosis
.
Leukemia
2024
;
38
:
610
20
.
108.
Schmitz
R
,
Wright
GW
,
Huang
DW
,
Johnson
CA
,
Phelan
JD
,
Wang
JQ
, et al
.
Genetics and pathogenesis of diffuse large B-cell lymphoma
.
N Engl J Med
2018
;
378
:
1396
407
.
109.
Chapuy
B
,
Stewart
C
,
Dunford
AJ
,
Kim
J
,
Kamburov
A
,
Redd
RA
, et al
.
Molecular subtypes of diffuse large B cell lymphoma are associated with distinct pathogenic mechanisms and outcomes
.
Nat Med
2018
;
24
:
679
90
.
110.
Locke
FL
,
Ghobadi
A
,
Jacobson
CA
,
Miklos
DB
,
Lekakis
LJ
,
Oluwole
OO
, et al
.
Long-term safety and activity of axicabtagene ciloleucel in refractory large B-cell lymphoma (ZUMA-1): a single-arm, multicentre, phase 1–2 trial
.
Lancet Oncol
2019
;
20
:
31
42
.
This open access article is distributed under the Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International (CC BY-NC-ND 4.0) license.