• Lenalidomide, obintuzumab, and CHOP chemotherapy are efficacious and well tolerated treatments for newly diagnosed DLBCL.

  • LO-CHOP led to high molecular response rates based on circulating tumor DNA sequencing during and after treatment in this phase 1b/2 study.

Diffuse large B-cell lymphoma (DLBCL) can be cured with rituximab, cyclophosphamide, doxorubicin, vincristine, and prednisone (R-CHOP); however, one-third of patients experience refractory or relapsed disease. Studies comparing R-CHOP with modified regimens replacing R with obinutuzumab (O) or adding lenalidomide (L) did not result in improved outcomes; however, L and O together may enhance natural killer-cell mediated antibody-dependent cellular toxicity when paired with CHOP. Here, we report on a phase 1b/2 study of 53 patients with newly diagnosed DLBCL who received 6 cycles of LO-CHOP. The end of treatment overall and complete response rates of the 50 evaluable patients were 98% and 90%, respectively. After a median follow-up of 4.5 years, the 4-year progression free and overall survival rates were 87.4% and 91.3%, respectively. Grade 3 to 4 adverse events were experienced by 70% of patients, including neutropenia (38%), thrombocytopenia (17%), fatigue (13%), and neutropenic fever (13%). Of the 33 patients profiled with circulating tumor DNA (ctDNA) sequencing, 31 (94%) had detectable pretreatment ctDNA with cancer personalized profiling by deep sequencing, 24 (73%) were classifiable by the LymphGen classifier, and 15/20 (75%) and 12/17 (71%) patients achieved early and major molecular responses after 1 and 2 cycles, respectively. Using phased variant enrichment and detection sequencing, 16/18 evaluable patients (89%) showed no detectable ctDNA after at least 5 cycles of LO-CHOP. LO-CHOP demonstrates high efficacy and tolerability in newly diagnosed DLBCL, leading to a high rate of undetectable minimal residual disease by ctDNA. This trial has been registered at www.clinicaltrials.gov as NCT02529852.

Diffuse large B-cell lymphoma (DLBCL) is the most common subtype of lymphoma,1 cured in approximately two-thirds of newly diagnosed patients with the immunochemotherapy regimen of rituximab, cyclophosphamide, doxorubicin, vincristine, and prednisone (R-CHOP).2,3 Multiple randomized studies investigating the addition of novel agents to the R-CHOP backbone have not definitively improved on R-CHOP as the prevailing standard-of-care frontline regimen4-8 before the recent POLARIX trial.9 The replacement of vincristine with the antibody-drug conjugate polatuzumab vedotin led to a modest progression free survival (PFS) improvement over R-CHOP for intermediate and high-risk disease, representing a shift in the preferred treatment of some patients.

Notable previous studies comparing R-CHOP to modified regimens include the GOYA trial (Obinutuzumab [O] with CHOP)6 and the ROBUST7 and Eastern Cooperative Oncology Group-ACRIN E14128 trials (lenalidomide [L] with R-CHOP). O is a glycoengineered type II antibody targeting CD20 that induces improved antibody-dependent cellular cytotoxicity, phagocytosis, and direct cell death compared with rituximab.10,11 The GOYA study was a randomized phase 3 trial that compared O-CHOP to R-CHOP in patients with untreated DLBCL. No difference was observed in the three-year PFS rates (70% vs 67%), response rates, or overall survival (OS), however, grade 3 to 5 adverse events (AEs) and discontinuation of any component of therapy were more common with O-CHOP.6 

L is an immunomodulatory agent that activates interferon signaling, inhibits the NF-κβ pathway,12 and promotes natural killer (NK) cell activity.13 In the setting of relapsed and refractory DLBCL, L monotherapy has shown activity, particularly for the activated B-cell (ABC) subtype.14 ROBUST was a phase 3 trial that randomized patients with treatment-naïve ABC DLBCL to LR-CHOP or R-CHOP. PFS and OS were not significantly different between the 2 arms; however, hematologic toxicities were more common with the addition of L. E1412 was a randomized phase 2 study that assigned patients with any subtype of untreated DLBCL to either LR-CHOP or R-CHOP. PFS was greater with LR-CHOP than with R-CHOP, with a hazard ratio of 0.67 and a 95% confidence interval (CI) of 0.44 to 1.03. The factors influencing the differential outcomes between these 2 trials may include a greater enrichment of high-risk patients in the E1412 study,15 differences in L administration, time to treatment, or other study population differences.

Neither O nor L alone have been adopted as part of frontline DLBCL therapy; however, the 2 agents may demonstrate synergistic activity when combined. The GALEN study investigated this combination in patients with relapsed or refractory follicular lymphoma16 or DLBCL,17 leading to overall response rates (ORR) of 81% and 35%, respectively. Excluding DLBCL patients with disease refractory to the last therapy or autologous stem cell transplant, the ORR was 61%, with a median PFS of 11.7 months. Patients treated with the combination experienced activation of NK cells and reversal of an immature NK cell phenotype as determined by cell surface markers.18 Administering the combination to newly diagnosed follicular lymphoma patients led to an ORR of 94%.19 We hypothesized that combining L and O with CHOP chemotherapy would demonstrate high efficacy with a tolerable toxicity profile. Herein, we report the results of a phase 1b/2 single center clinical trial (NCT02529852) evaluating LO-CHOP for the treatment of patients with newly diagnosed DLBCL. Circulating tumor DNA (ctDNA) profiling was performed to measure the baseline tumor burden, molecular response, and molecular subtypes.

Study design

This investigator initiated, open label, single-arm phase 1b/2 study was conducted at the University of Texas MD Anderson Cancer Center. The primary end point of the phase Ib portion was to evaluate the safety of LO-CHOP and the primary end point of the phase 2 portion was to evaluate the clinical efficacy of LO-CHOP at the end of therapy. The safety evaluation in phase 1b was defined as the lack of any grade ≥3 nonhematologic toxicity unmanageable with aggressive supportive care or toxicity, resulting in a delay of over 7 days of cycle 2. The efficacy evaluation in phase 2 was defined as the ORR and complete response rate (CRR) at the end of therapy assessment with positron emission tomography/computed tomography (PET/CT) imaging, as defined by the Lugano 2014 classification.20 Secondary endpoints for the study included rates of AEs, PFS, and OS. PFS was defined as the time from the start of therapy until the date of disease progression or death as a result of any cause and censored at the last follow-up date in disease remission, and OS was defined as the time from the start of therapy until the date of death as a result of any cause and censored at the last follow-up date while alive. AEs were defined according to the National Cancer Institute Common Terminology Criteria for Adverse Events v4.0. This study was approved by the Institutional Review Board of the MD Anderson Cancer Center under protocol 2015-0069 and conducted in accordance with the Declaration of Helsinki.

Key eligibility criteria

Eligible patients were aged ≥18 years with newly diagnosed untreated CD20+ DLBCL. Patients with stage I DLBCL were eligible only if 6 cycles of chemotherapy were planned. Measurable disease of at least 1.5 cm in longest diameter on imaging was required, as was an Eastern Cooperative Oncology Group performance status of 2 or less, unless the previously performance status was 0 to 1 and deterioration was felt to be due to lymphoma and reversible with therapy. Patients were required to have adequate organ and bone marrow function. Patients were ineligible if they were pregnant or nursing women, had a prior diagnosis of low-grade lymphoma, had central nervous system involvement, active HIV infection, active Hepatitis B or C infection, or were unwilling to take aspirin for venous thrombosis prophylaxis.

Pathology review

All diagnostic tumor biopsies were reviewed by expert hematopathologists at the University of Texas MD Anderson Cancer Center for confirmation of DLBCL using the standard WHO diagnostic criteria. The cell of origin (COO) was determined via immunohistochemistry (IHC) using the Hans algorithm.21 When adequate tissue was available for standard testing, double expressor lymphoma was defined as positivity by IHC staining for MYC and BCL2, and double-hit lymphoma was defined as DLBCL with a rearrangement of MYC with concurrent rearrangement of BCL2 and/or BCL6 by fluorescence in situ hybridization.

Treatment

Patients were treated with L 15 mg orally daily on days 1 through 14 of each 21-day cycle, O 1000 mg intravenously on days 1, 8, and 15 of cycle 1 and day 1 only for all additional cycles, and standard CHOP (cyclophosphamide 750 mg/m2, doxorubicin 50 mg/m2, vincristine 1.4 mg/m2 [capped at 2.0 mg], and prednisone 100 mg by mouth daily on days 1 through 5) starting on day 1 of all cycles. Hematologic AEs that required dose reduction of L were recurrent grade 3 neutropenia (sustained for ≥7 days or with fever), recurrent grade 4 neutropenia, or recurrent grade ≥3 thrombocytopenia. All patients were scheduled to receive 6 cycles if no progression occurred, and no maintenance therapy was administered. All patients were required to receive granulocyte colony stimulating factor, aspirin 81 mg if not already receiving anticoagulation, and Pneumocystis jirovecii pneumonia prophylaxis therapy. Intrathecal chemotherapy was allowed if the treating physician deemed the patient at risk for central nervous system involvement because of standard risk factors.22 

ctDNA profiling

Plasma samples were collected in EDTA tubes. Cell-free DNA (cfDNA) was extracted from 1 to 5 mL of plasma using a Qiagen QIAamp Circulating Nucleic Acid Kit. Somatic alterations were called from cfDNA using cancer personalized profiling by deep sequencing, as previously described.23-26 Hybrid-capture was performed using targeted panels optimized for B-cell lymphomas. Quantitative levels of ctDNA were measured in haploid genome equivalents per milliliter (hGE/mL), determined as the product of total cfDNA concentration and the mean allele fraction of somatic mutations, expressed in log scale (log hGE/mL), as previously reported.25,27 Patients with ≥20 coding and silent mutations detected by noninvasive genotyping were subjected to genotypic classification using the LymphGen online tool (https://llmpp.nih.gov/lymphgen/index.php).28 For this purpose, mutations were integrated with genome-wide copy number alterations called from on- and off-target sequencing reads as previously described.26 

We applied “phased variant enrichment and detection sequencing” (PhasED-Seq) to track minimal residual disease (MRD) in on-treatment samples29 from patients with available baseline samples. Patients were stratified using previously established molecular milestones, where early molecular response (EMR) was defined as a 2 log drop in ctDNA after 1 cycle of therapy, and major molecular response (MMR) as a 2.5 log drop in ctDNA after 2 cycles of therapy.25 

Statistical considerations

Descriptive statistics were used to define baseline characteristics and treatment response/outcomes. Fisher exact test and Wilcoxon rank-sum test were used to evaluate the association between the end of therapy response with categorical variables and continuous variables, respectively. The Kaplan-Meier method was used to estimate the probabilities of PFS/OS, and the log-rank test was used to evaluate differences in PFS/OS. The cox proportional hazards model was used to estimate the hazard ratio for the covariates. Statistical software used included SAS 9.4 (SAS Institute Inc, Cary, NC), S-Plus 8.2 (TIBCO Software Inc, Palo Alto, CA), and R 4.1.2 (R Core Team, Vienna, Austria).

Patients

Fifty-three patients were enrolled, 6 in the phase 1b portion and 47 in the phase 2 portion. Patients started treatment between 12 November 2015 and 30 March 2017. The median patient age was 62 years (range 26-83), 35 (66%) had stage III/IV disease, 26 (49%) had elevated lactate dehydrogenase, 12 (23%) had an international prognostic index (IPI) >2, 26 (49%) had germinal center (GC), 22 (42%) had non-GC subtype, and 5 were unable to be subtyped. Detailed baseline characteristics are summarized in Table 1.

Table 1.

Baseline patient characteristics

CharacteristicsAll patients [N = 53], N (%)
Median age, y (range) 62 (26-83) 
Age >60 y 28 (53) 
Male sex 30 (57) 
Ann Arbor stage  
2 (4) 
II 16 (30) 
III 18 (34) 
IV 17 (32) 
Extranodal sites >1 4 (8) 
ECOG performance status  
24 (45) 
27 (51) 
2 (4) 
Lactate dehydrogenase > upper limit normal 26 (49) 
IPI  
2 (4) 
1-2 39 (74) 
3-4 12 (23) 
Bulky disease >10 cm 5 (9) 
Double expressor lymphoma (N = 32) 9 (28) 
Double hit lymphoma (N = 34) 3 (9) 
Cell-of-origin  
GCB 26 (49) 
Non-GCB 22 (42) 
NA 5 (9) 
CharacteristicsAll patients [N = 53], N (%)
Median age, y (range) 62 (26-83) 
Age >60 y 28 (53) 
Male sex 30 (57) 
Ann Arbor stage  
2 (4) 
II 16 (30) 
III 18 (34) 
IV 17 (32) 
Extranodal sites >1 4 (8) 
ECOG performance status  
24 (45) 
27 (51) 
2 (4) 
Lactate dehydrogenase > upper limit normal 26 (49) 
IPI  
2 (4) 
1-2 39 (74) 
3-4 12 (23) 
Bulky disease >10 cm 5 (9) 
Double expressor lymphoma (N = 32) 9 (28) 
Double hit lymphoma (N = 34) 3 (9) 
Cell-of-origin  
GCB 26 (49) 
Non-GCB 22 (42) 
NA 5 (9) 

ECOG, Eastern Cooperative Oncology Group; GCB, germinal center B-cell; N, number; NA, not applicable.

Safety

Treatment related AEs occurred in all patients, and 37 (70%) patients experienced at least 1 grade 3 to 4 treatment related AE. None of the patients experienced fatal AE. There were no dose limiting toxicities for the 6 patients enrolled in the phase I∖1b portion at a maximum tolerated dose of L at 15 mg. Common any grade AEs included fatigue (96%), constipation (85%), nausea (72%), anemia (62%), myalgia (59%), and dizziness (57%). Common grade 3 to 4 AEs included neutropenia (38%), thrombocytopenia (17%), fatigue (13%), febrile neutropenia (13%), and infection (9%). Grades 3 to 4 infections included cellulitis, urinary tract infection, pneumonia, and Clostridium difficile colitis. Four patients (8%) experienced thromboembolism. The AEs are summarized in Table 2.

Table 2.

Summary of grade 3 to 4 treatment emergent AEs (≥5%) and AEs of interest

AEAny gradeGrade 3-4
Most common AEs   
Neutropenia 21 (40) 20 (38) 
Thrombocytopenia 15 (28) 9 (17) 
Fatigue 51 (96) 7 (13) 
Febrile neutropenia 7 (13) 7 (13) 
Infection 12 (23) 5 (9) 
Myalgia 31 (59) 4 (8) 
Pain 18 (34) 3 (6) 
AEs of interest   
Constipation 45 (85) 1 (2) 
Nausea 38 (72) 2 (4) 
Anemia 33 (62) 1 (2) 
Peripheral sensory neuropathy 22 (42) 1 (2) 
Diarrhea 21 (40) 1 (2) 
Venous thromboembolism 4 (8) 0 (0) 
AEAny gradeGrade 3-4
Most common AEs   
Neutropenia 21 (40) 20 (38) 
Thrombocytopenia 15 (28) 9 (17) 
Fatigue 51 (96) 7 (13) 
Febrile neutropenia 7 (13) 7 (13) 
Infection 12 (23) 5 (9) 
Myalgia 31 (59) 4 (8) 
Pain 18 (34) 3 (6) 
AEs of interest   
Constipation 45 (85) 1 (2) 
Nausea 38 (72) 2 (4) 
Anemia 33 (62) 1 (2) 
Peripheral sensory neuropathy 22 (42) 1 (2) 
Diarrhea 21 (40) 1 (2) 
Venous thromboembolism 4 (8) 0 (0) 

Because of AEs, 23 (43%) patients experienced L interruption (mainly owing to midcycle neutropenia or thrombocytopenia), 11 (21%) required a dose reduction of L, and 3 (6%) discontinued L. One patient required a delay of >7 days in their next cycle of LO-CHOP. The reasons for dose reduction were as follows: recurrent grade ≥3 neutropenia in 6 patients, and 1 patient each experienced a single episode of concurrent grade ≥3 neutropenia and thrombocytopenia, a single episode of febrile neutropenia, colitis, hospitalization for weakness, and recurrent infections. Of the 3 patients who discontinued L for AEs, 1 patient discontinued after 5 cycles and still completed a sixth cycle of O-CHOP, 1 patient came off the study after 3 cycles and completed another 3 cycles of R-CHOP without vincristine, and 1 patient discontinued therapy after 1 cycle.

Efficacy

Fifty-one patients were evaluated using interim PET/CT after 2 cycles of LO-CHOP (2 patients could not be evaluated because of study withdrawal, 1 because of AEs, and 1 per patient preference). The ORR was 51/51 (100%) at the interim assessment and the CRR was 43/51 (84%; 95% CI, 71-93) whereas the partial response rate was 8/51 (16%). Fifty patients were evaluated at the end of therapy after 6 cycles of LO-CHOP with PET/CT (an additional patient withdrew because of AEs). The ORR at the end of therapy was 49/50 (98%; 95% CI, 89-100) and 45/50 (90%; 95% CI, 78-97), 4 (8%), and 1 (2%) patient experienced CR, partial response, or progressive disease, respectively.

After a median follow-up of 4.5 years (range 1.1-5.8), 6 patients experienced disease progression or relapse, and 4 patients died. The median PFS and OS were not reached, and the 4-year PFS and OS rates were 87.4% (95% CI, 78.4-97.5) and 91.3% (95% CI, 83.5-99.9), respectively (Figure 1A-B). Two patients received high dose chemotherapy, followed by autologous stem cell transplant after progression. No baseline patient or disease characteristics were associated with the ORR, CRR, PFS, or OS. The causes of death were progressive lymphoma, transformed myelodysplastic syndrome, cerebrovascular accident, and unknown.

Figure 1.

Time-to-event survival outcomes. PFS (A) and OS (B) of the entire cohort.

Figure 1.

Time-to-event survival outcomes. PFS (A) and OS (B) of the entire cohort.

Close modal

ctDNA profiling

Baseline plasma samples were retrieved from of 33/53 patients (62%). A total of 31/33 (94%) of the profiled patients had baseline ctDNA detectable with cancer personalized profiling by deep sequencing. The median baseline log ctDNA concentration was 2.02 log hGE/mL (range 0-3.29), and 8/33 (24%) of the patients had high baseline ctDNA levels by a previously established threshold of 2.5 log hGE/mL.25,27 Nine patients were not evaluable for genotyping and molecular response assessment owing to low pretreatment ctDNA levels, which, in the absence of matched tumor tissue, precluded the identification of sufficient reporters. Of these, only 1 patient experienced a PFS event during follow-up at 3.8 years, reflective of the overall favorable risk profile of patients with low levels of pretreatment ctDNA. A total of 24/33 (73%) patients were successfully evaluated using the LymphGen classifier. Molecular subtypes, in order of decreasing prevalence, included other (9, 38%), EZB (5, 21%), ST2 (5, 21%), MCD (4, 17%), and A53 (1, 4%) (Figure 2A). Two patients had composite subtypes (EZB/ST2 and EZB/A53), and were classified as EZB here. As expected, all 5 EZB and 4/5 ST2-classified patients were classified as having the GC subtype, whereas only 2/4 MCD patients were non-GC according to the Hans algorithm. The genotypes of the 2 remaining patients with MCD were manually reviewed, and their classification as MCD was deemed appropriate based on the presence of canonical hotspot mutations, suggesting that these cases may have been misclassified as COO by IHC. The prevalence of COO subgroups within each genetic subtype is shown in Figure 2B.

Figure 2.

Description of molecular subtypes by LymphGen classifier. Frequency of molecular subtypes (A). Prevalence of COO subtypes within molecular subtypes (B).

Figure 2.

Description of molecular subtypes by LymphGen classifier. Frequency of molecular subtypes (A). Prevalence of COO subtypes within molecular subtypes (B).

Close modal

The samples used to evaluate EMR and MMR were available for 20 and 17 of the 24 eligible patients, respectively. The EMR and MMR rates were 15/20 (75%) and 12/17 (71%) (Figure 3A-B). Among the 15 patients evaluable for both EMR and MMR, the concordance between the 2 endpoints was 13/15 (87%). The fold change in ctDNA levels through all time points is depicted in Figure 3C. Of the 3 profiled cases with progression/relapse events, 1 did not achieve EMR and MMR, 1 initially cleared their ctDNA at the EMR and MMR time points but had detectable ctDNA after cycle 3, and 1 did not have detectable pretreatment ctDNA. These patients did not have the end of treatment samples available. A total of 18 patients were evaluated for ctDNA assessment after at least 5 cycles of therapy. Of these, 16 (89%) were undetectable for ctDNA by PhasED-seq, whereas 2 patients had detectable ctDNA at this late time point; one of these patients was diagnosed with a previously unknown concurrent localized follicular lymphoma after the end of therapy, which was successfully treated with radiation alone, and had not experienced relapse of DLBCL or follicular lymphoma. The other patient remained in CR with 55 months of follow-up after achieving EMR, with a 700-fold reduction in the tumor burden during LO-CHOP. No ctDNA-based characteristics, including molecular subtype, EMR, MMR, or late time point MRD by PhasED-Seq, were associated with significant differences in response rates, PFS, or OS in the profiled patients.

Figure 3.

ctDNA-defined molecular response dynamics. Waterfall plots depicting the log-fold change in ctDNA levels from pretreatment to after cycle 1 (A) and after cycle 2 (B). Bars are colored in accordance with the end-of-therapy response. Dashed lines signify the EMR (2 log-fold decrease) and MMR (2.5 log-fold decrease) thresholds. Spider plot (C) depicting ctDNA level dynamics across all time points when the samples were available. Lines are colored in accordance with disease status at time of data censoring. The dashed lines signify the EMR and MMR thresholds at their respective time points. ND, not detected.

Figure 3.

ctDNA-defined molecular response dynamics. Waterfall plots depicting the log-fold change in ctDNA levels from pretreatment to after cycle 1 (A) and after cycle 2 (B). Bars are colored in accordance with the end-of-therapy response. Dashed lines signify the EMR (2 log-fold decrease) and MMR (2.5 log-fold decrease) thresholds. Spider plot (C) depicting ctDNA level dynamics across all time points when the samples were available. Lines are colored in accordance with disease status at time of data censoring. The dashed lines signify the EMR and MMR thresholds at their respective time points. ND, not detected.

Close modal

The combination of L and O with CHOP chemotherapy in this phase 1b/2 study for the treatment of newly diagnosed DLBCL was highly efficacious, leading to end of therapy overall and CR rates of 96% and 90%, and 4-year PFS and OS rates of 87% and 91%, respectively. LO-CHOP was similarly efficacious in high-risk patient subsets, including those with elevated IPI, non-GCB COO, high pretreatment ctDNA levels, and MCD and EZB molecular subtypes.28 Rates of EMR and MMR after 1 or 2 cycles of LO-CHOP were 75% and 71%, respectively, with high concordance between these 2 endpoints in patients with both assessments performed. Encouragingly, end of therapy assessment with PhasED-seq demonstrated a high rate of undetectable MRD (89%), with 1 detectable patient being diagnosed with indolent lymphoma after therapy. Here, we demonstrate that targeted ctDNA sequencing allows for noninvasive molecular classification of patients treated in a prospective clinical trial by genetic subtype.

The safety profile of LO-CHOP did not result in any unexpected toxicities; with mandated growth factor support, the rate of grade 3 to 4 AEs, such as neutropenia, thrombocytopenia, and febrile neutropenia, compared favorably to those seen in LR-CHOP and O-CHOP treated patients.6-8 The rate of any-grade thromboembolism may have been higher than that in the ROBUST study, although it is difficult to draw conclusions with small patient numbers. The rates of AEs of any grade, such as fatigue, nausea, and constipation, were higher than expected, but a lack of dose limiting toxicities, low rate of treatment discontinuation or delay, and similar incidence of cytopenias compared with other studies suggest that this discrepancy could be related to differences in reporting rather than excessive toxicity compared with R-CHOP or LR-CHOP.

The limitations of this study stemmed from its single-arm design. Without a comparator arm, we could not determine whether LO-CHOP could improve the efficacy compared with R-CHOP. The low number of progression events precluded the analysis of baseline or molecular characteristics, leading to the differential efficacy of LO-CHOP, including ctDNA sequencing. Only 3 patients who experienced disease progression had ctDNA profiled. Furthermore, our cohort was a relatively low-risk population with a lower proportion of patients with negative prognostic features such as advanced stage, elevated lactate dehydrogenase, extranodal sites, or IPI score.

The GOYA, E1412, and ROBUST studies did not support the replacement of R-CHOP as the standard treatment for newly diagnosed DLBCL. Other randomized studies evaluating the additive benefits of L in frontline therapy include the REMARC30 and SENIOR31 studies. The former found that L maintenance after R-CHOP in elderly patients improved PFS but not OS, whereas the latter demonstrated that the addition of L to dose-reduced R-CHOP (R-miniCHOP) in elderly patients did not improve PFS or OS. Despite the high-efficacy of LO-CHOP in this study, based on the published results of other trials, it appears that L and O, on their own or in combination, may still be better reserved for the treatment of indolent lymphomas.16,19,32-35 Encouraging single-arm phase 2 results when adding novel agents to R-CHOP has not been translated into positive phase 3 studies in most cases, potentially because of patient selection pitfalls and failure to enrich high-risk patients.36,37 Although the patients treated in the current study achieved excellent outcomes, it is not clear how this would have differed from treatment with R-CHOP.

Emerging evidence suggests that transcriptionally and genetically defined DLBCL subtypes do not only have prognostic and predictive significances. Although the PFS improvement with the use of polatuzumab vedotin over vincristine was only 6.5% in the full POLARIX study cohort, the effect was more pronounced in the ABC subset.9 Similarly, MCD and BN2 subtypes may be more susceptible to transcriptional changes induced by L or B-cell receptor signaling inhibition through ibrutinib.28,38 Of note, none of the 4 MCD patients in this study progressed. In the future, treatment may be guided by molecular profiling.

Although studies targeting molecular subgroups to date have uniformly utilized tumor tissue for this purpose,4,5,7,38 liquid biopsies have several advantages. For example, liquid biopsies could facilitate molecularly-guided studies, given their noninvasive character, better capture of tumor heterogeneity, and allow access to abundant fresh, nonfixed specimens exclusively dedicated to molecular analyses. Separately, molecular response assessment using ctDNA may prove to be an effective early surrogate end point to uncover differences in efficacy and lead to accelerated decision making and drug approval.39 

A limitation of studies abstaining from tumor tissue is that sufficient levels of pretreatment ctDNA are needed for the de novo identification of somatic alterations. Specifically, ctDNA assays typically require variant allele frequencies of ≥0.5% to reliably call mutations.40 This is especially relevant for lower-risk cohorts, such as those in which only 24% of profiled patients had high ctDNA levels, compared with ∼40% to 50% in previous cohorts.25,27 Therefore, a fully noninvasive approach carries the risk of excluding cases with low baseline ctDNA levels from downstream analyses, which may ultimately require the integration of plasma and tumor sequencing in a subset of cases.

Leveraging synergistic combinations of novel agents to exploit synthetic lethality12 may be more effective than adding “X” novel agent alone to R-CHOP if additional toxicity can be managed. Promising combination regimens include tafasitamab and L with R-CHOP41,42 and ibrutinib and L with R-CHOP.43 

In summary, LO-CHOP as a modified frontline regimen for DLBCL in this single-arm phase 1b/2 study showed high rates of radiographic and molecular responses, durable remission with long-term follow-up, and an expected safety profile. The design approaches included in this study, including novel-novel combinations and ctDNA-based genetic classification, may be essential to further improve frontline therapy in DLBCL.

This study was funded by the MD Anderson National Institutes of Health/National Cancer Institute Cancer Support Grant under award number P30 CA016672, and R01CA233975 (A.A.A.). M.R.G. is a special fellow of the Leukemia & Lymphoma Society. A.A.A. is a scholar of the Leukemia & Lymphoma Society.

Contribution: H.-J.J.C., S.K.A., A.F.M.C., and C.W.M. collected and analyzed data; H.-J.J.C. and S.A. wrote the manuscript; Y.O., L.J.N., L.F., S.S.N., F.T., F.H., M.A.R., H.J.L., and C.R.F. cared for patients and critically reviewed the manuscript; S.K.A., A.F.M.C., C.W.M., M.R.G., A.A.A., and R.E.D. performed experiments; T.J.M. and F.V. performed histopathologic analysis; L.F. analyzed data; J.R.W. designed the study, collected and analyzed data, and wrote the manuscript; and all authors approved the final version of the manuscript.

Conflict-of-interest disclosure: S.K.A. reports speaker honoraria from Takeda. L.J.N. received honorarium from ADC Therapeutics, Bristol Myers Squibb (BMS), Epizyme, Genentech/Roche, Genmab, Janssen, MorphoSys, Novartis, Takeda, and research support from BMS, Caribou Biosciences, Epizyme, Genentech, Genmab, IGM Biosciences, Janssen, Novartis, and Takeda. S.S.N. received research support from Kite/Gilead, BMS, Cellectis, Poseida, Allogene, Unum Therapeutics, Precision Biosciences, and Adicet Bio; served as an advisory board member/consultant for Kite/Gilead, Merck, Novartis, Sellas Life Sciences, Athenex, Allogene, Incyte, Adicet Bio, BMS, Legend Biotech, bluebird bio, Sana Biotechnology, Caribou Biosciences, and Astellas Pharma; has received royalty income from Takeda Pharmaceuticals; has stock options from Longbow Immunotherapy, Inc; and has intellectual property related to cell therapy. C.R.F. reports consultancy for AstraZeneca, Bayer, BeiGene, BioAscend, BMS, Celgene, Curio Sciences, Denovo Biopharma, Epizyme/Incyte, Foresight Diagnostics, Genentech/Roche, Genmab, MEI Pharmaceuticals, MorphoSys AG, Pharmacyclics/Janssen, SeaGen, and research funding from 4D, AbbVie, Acerta, Adaptimmune, Allogene, Amgen, Bayer, Celgene, Cellectis, EMD, Gilead, Genentech/Roche, Guardant, Iovance, Janssen Pharmaceutical, Kite, MorphoSys, Nektar, Novartis, Pfizer, Pharmacyclics, Sanofi, Takeda, TGTherapeutics, Xencor, Ziopharm, Burroughs Wellcome Fund, Eastern Cooperative Oncology Group, National Cancer Institute, V Foundation, Cancer Prevention and Research Institute of Texas: CPRIT Scholar in Cancer Research. F.V. receives research funding from CRISPR Therapeutics, Allogene, and Geron Corporation. M.R.G. reports research funding from Sanofi, Kite/Gilead, AbbVie, and Allogene, honoraria from Tessa Therapeutics, Monte Rosa Therapeutics, and Daiichi Sankyo, and stock ownership of KDAc Therapeutics. D.M.K. reports ownership interest in Foresight Diagnostics, personal fees from Roche, and patent filings related to cancer biomarkers. A.A.A. reports personal fees and other support from Roche, Gilead, and Chugai, other support from Genentech, Celgene, Janssen, FortySeven, Foresight Diagnostics, CiberMed, and Pharmacyclics, and personal fees and other support from grants from BMS during the conduct of the study. J.R.W. has research funding from Kite/Gilead, BMS, Novartis, Genentech/Roche, MorphoSys/Incyte, AstraZeneca, ADC Therapeutics, and consulting funding for Kite/Gilead, BMS, Novartis, Genentech/Roche, MorphoSys/Incyte, AstraZeneca, ADC Therapeutics, Merck, MonteRosa, Umoja, and Ikusda. The remaining authors declare no competing financial interests.

Correspondence: Jason R. Westin, Department of Lymphoma & Myeloma, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Unit 429, Houston, TX 77030; e-mail: jwestin@mdanderson.org.

1.
Swerdlow
SH
,
Campo
E
,
Harris
NL
, et al
.
WHO Classification of Tumors of Haematopoietic and Lymphoid Tissues
. Revised 4th.
International Agency for Research on Cancer
;
2017
. .
2.
Coiffier
B
,
Thieblemont
C
,
van den Neste
E
, 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
(
12
):
2040
-
2045
.
3.
Pfreundschuh
M
,
Kuhnt
E
,
Trümper
L
, et al
.
CHOP-like chemotherapy with or without rituximab in young patients with good-prognosis diffuse large-B-cell lymphoma: 6-year results of an open-label randomised study of the MabThera International Trial (MInT) Group
.
Lancet Oncol
.
2011
. ;
12
(
11
):
1013
-
1022
.
4.
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
(
15
):
1285
-
1295
.
5.
Davies
A
,
Cummin
TE
,
Barrans
S
, et al
.
Gene-expression profiling of bortezomib added to standard chemoimmunotherapy for diffuse large B-cell lymphoma (REMoDL-B): an open-label, randomised, phase 3 trial
.
Lancet Oncol
.
2019
. ;
20
(
5
):
649
-
662
.
6.
Vitolo
U
,
Trneny
M
,
Belada
D
, et al
.
Obinutuzumab or rituximab plus cyclophosphamide, doxorubicin, vincristine, and prednisone in previously untreated diffuse large b-cell lymphoma
.
J Clin Oncol
.
2017
. ;
35
(
31
):
3529
-
3537
.
7.
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
(
12
):
1317
-
1328
.
8.
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
(
12
):
1329
-
1338
.
9.
Tilly
H
,
Morschhauser
F
,
Sehn
LH
, et al
.
Polatuzumab vedotin in previously untreated diffuse large B-cell lymphoma
.
N Engl J Med
.
2022
. ;
386
(
4
):
351
-
363
.
10.
Mössner
E
,
Brünker
P
,
Moser
S
, et al
.
Increasing the efficacy of CD20 antibody therapy through the engineering of a new type II anti-CD20 antibody with enhanced direct and immune effector cell-mediated B-cell cytotoxicity
.
Blood
.
2010
. ;
115
(
22
):
4393
-
4402
.
11.
Herter
S
,
Herting
F
,
Mundigl
O
, et al
.
Preclinical activity of the type II CD20 antibody GA101 (obinutuzumab) compared with rituximab and ofatumumab in vitro and in xenograft models
.
Mol Cancer Ther
.
2013
. ;
12
(
10
):
2031
-
2042
.
12.
Yang
Y
,
Shaffer
AL
,
Emre
NCT
, et al
.
Exploiting synthetic lethality for the therapy of ABC diffuse large B cell lymphoma
.
Cancer Cell
.
2012
. ;
21
(
6
):
723
-
737
.
13.
Acebes-Huerta
A
,
Huergo-Zapico
L
,
Gonzalez-Rodriguez
AP
, et al
.
Lenalidomide induces immunomodulation in chronic lymphocytic leukemia and enhances antitumor immune responses mediated by NK and CD4 T cells
.
BioMed Res Int
.
2014
. ;
2014
:
265840
.
14.
Czuczman
MS
,
Trněný
M
,
Davies
A
, et al
.
A phase 2/3 multicenter, randomized, open-label study to compare the efficacy and safety of lenalidomide versus investigator’s choice in patients with relapsed or refractory diffuse large B-cell lymphoma
.
Clin Cancer Res
.
2017
. ;
23
(
15
):
4127
-
4137
.
15.
Nowakowski
G
,
Chiappella
A
,
Hong
F
, et al
.
Potential factors that impact lenalidomide/R-CHOP efficacy in previously untreated diffuse large B-cell lymphoma in the ROBUST and ECOG-ACRIN 1412 studies
.
Blood
.
2019
. ;
134
(
suppl 1
):
4092
.
16.
Morschhauser
F
,
le Gouill
S
,
Feugier
P
, et al
.
Obinutuzumab combined with lenalidomide for relapsed or refractory follicular B-cell lymphoma (GALEN): a multicentre, single-arm, phase 2 study
.
Lancet Haematol
.
2019
. ;
6
(
8
):
e429
-
e437
.
17.
Houot
R
,
Cartron
G
,
Bijou
F
, et al
.
Obinutuzumab plus lenalidomide (GALEN) for the treatment of relapse/refractory aggressive lymphoma: a phase II LYSA study
.
Leukemia
.
2019
. ;
33
(
3
):
776
-
780
.
18.
Vo
DN
,
Alexia
C
,
Allende-Vega
N
, et al
.
NK cell activation and recovery of NK cell subsets in lymphoma patients after obinutuzumab and lenalidomide treatment
.
OncoImmunology
.
2017
. ;
7
(
4
):
e1409322
.
19.
Bachy
E
,
Houot
R
,
Feugier
P
, et al
.
Obinutuzumab plus lenalidomide in advanced, previously untreated follicular lymphoma in need of systemic therapy: a LYSA study
.
Blood
.
2022
. ;
139
(
15
):
2338
-
2346
.
20.
Cheson
BD
,
Fisher
RI
,
Barrington
SF
, et al
.
Recommendations for initial evaluation, staging, and response assessment of Hodgkin and non-Hodgkin lymphoma: the Lugano classification
.
JJ Clin Oncol
.
2014
. ;
32
(
27
):
3059
-
3067
.
21.
Hans
CP
,
Weisenburger
DD
,
Greiner
TC
, et al
.
Confirmation of the molecular classification of diffuse large B-cell lymphoma by immunohistochemistry using a tissue microarray
.
Blood
.
2004
. ;
103
(
1
):
275
-
282
.
22.
Schmitz
N
,
Zeynalova
S
,
Nickelsen
M
, et al
.
CNS International Prognostic Index: a risk model for CNS relapse in patients with diffuse large B-cell lymphoma treated with R-CHOP
.
J Clin Oncol
.
2016
. ;
34
(
26
):
3150
-
3156
.
23.
Newman
AM
,
Bratman
S V
,
To
J
, et al
.
An ultrasensitive method for quantitating circulating tumor DNA with broad patient coverage
.
Nat Med
.
2014
. ;
20
(
5
):
548
-
554
.
24.
Newman
AM
,
Lovejoy
AF
,
Klass
DM
, et al
.
Integrated digital error suppression for improved detection of circulating tumor DNA
.
Nat Biotechnol
.
2016
. ;
34
(
5
):
547
-
555
.
25.
Kurtz
DM
,
Scherer
F
,
Jin
MC
, et al
.
Circulating tumor DNA measurements as early outcome predictors in diffuse large B-cell lymphoma
.
J Clin Oncol
.
2018
. ;
36
(
28
):
2845
-
2853
.
26.
Chabon
JJ
,
Hamilton
EG
,
Kurtz
DM
, et al
.
Integrating genomic features for non-invasive early lung cancer detection
.
Nature
.
2020
. ;
580
(
7802
):
245
-
251
.
27.
Alig
S
,
Macaulay
CW
,
Kurtz
DM
, et al
.
Short diagnosis-to-treatment interval is associated with higher circulating tumor DNA levels in diffuse large B-cell lymphoma
.
J Clin Oncol
.
2021
. ;
39
(
23
):
2605
-
2616
.
28.
Wright
GW
,
Huang
DW
,
Phelan
JD
, et al
.
A probabilistic classification tool for genetic subtypes of diffuse large B cell lymphoma with therapeutic implications
.
Cancer Cell
.
2020
. ;
37
(
4
):
551
-
568.e14
.
29.
Kurtz
DM
,
Soo
J
,
Co Ting Keh
L
, et al
.
Enhanced detection of minimal residual disease by targeted sequencing of phased variants in circulating tumor DNA
.
Nat Biotechnol
.
2021
. ;
39
(
12
):
1537
-
1547
.
30.
Thieblemont
C
,
Tilly
H
,
da Silva
MG
, et al
.
Lenalidomide maintenance compared with placebo in responding elderly patients with diffuse large B-cell lymphoma treated with first-line rituximab plus cyclophosphamide, doxorubicin, vincristine, and prednisone
.
J Clin Oncol
.
2017
. ;
35
(
22
):
2473
-
2481
.
31.
Oberic
L
,
Peyrade
F
,
Puyade
M
, et al
.
Subcutaneous rituximab-MiniCHOP compared with subcutaneous rituximab-MiniCHOP plus lenalidomide in diffuse large B-cell lymphoma for patients age 80 years or older
.
J Clin Oncol
.
2021
. ;
39
(
11
):
1203
-
1213
.
32.
Marcus
R
,
Davies
A
,
Ando
K
, et al
.
Obinutuzumab for the first-line treatment of follicular lymphoma
.
N Engl J Med
.
2017
. ;
377
(
14
):
1331
-
1344
.
33.
Sehn
LH
,
Chua
N
,
Mayer
J
, et al
.
Obinutuzumab plus bendamustine versus bendamustine monotherapy in patients with rituximab-refractory indolent non-Hodgkin lymphoma (GADOLIN): a randomised, controlled, open-label, multicentre, phase 3 trial
.
Lancet Oncol
.
2016
. ;
17
(
8
):
1081
-
1093
.
34.
Leonard
JP
,
Trneny
M
,
Izutsu
K
, et al
.
AUGMENT: a phase III study of lenalidomide plus rituximab versus placebo plus rituximab in relapsed or refractory indolent lymphoma
.
J Clin Oncol
.
2019
. ;
37
(
14
):
1188
-
1199
.
35.
Morschhauser
F
,
Fowler
NH
,
Feugier
P
, et al
.
Rituximab plus lenalidomide in advanced untreated follicular lymphoma
.
N Engl J Med
.
2018
. ;
379
(
10
):
934
-
947
.
36.
Cherng
HJJ
,
Westin
J
.
Why R-CHOP + X is not enough: lessons learned and next steps in the mission to improve frontline therapy for diffuse large B-cell lymphoma
.
Leuk Lymphoma
.
2021
. ;
62
(
6
):
1302
-
1312
.
37.
Lue
JK
,
O’Connor
OA
.
A perspective on improving the R-CHOP regimen: from Mega-CHOP to ROBUST R-CHOP, the PHOENIX is yet to rise
.
Lancet Haematol
.
2020
. ;
7
(
11
):
e838
-
e850
.
38.
Wilson
WH
,
Wright
GW
,
Huang
DW
, et al
.
Effect of ibrutinib with R-CHOP chemotherapy in genetic subtypes of DLBCL
.
Cancer Cell
.
2021
. ;
39
(
12
):
1643
-
1653.e3
.
39.
Food and Drug Administration
.
Use of circulating tumor DNA for early-stage solid tumor drug development guidance for industry
. Accessed 25 May 2022. https://www.fda.gov/media/158072/download.
40.
Deveson
IW
,
Gong
B
,
Lai
K
, et al
.
Evaluating the analytical validity of circulating tumor DNA sequencing assays for precision oncology
.
Nat Biotechnol
.
2021
. ;
39
(
9
):
1115
-
1128
.
41.
Belada
D
,
Kopeckova
K
,
Bergua
JM
, et al
.
First-MIND: a phase Ib, open-label, randomized study to assess safety of tafasitamab (tafa) or tafa + lenalidomide (LEN) in addition to R-CHOP in patients with newly diagnosed DLBCL
.
J Clin Oncol
.
2021
. ;
39
(
15 suppl
):
7540
.
42.
Vitolo
U
,
Nowakowski
GS
,
Burke
JM
, 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 (DLBCL)
.
Clin Lymphoma Myeloma Leuk
.
2021
. ;
21
:
S376
-
S377
.
43.
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
.
Published online 11 August 2022
. .

Author notes

H.-J.J.C. and S.K.A. contributed equally to this study.

Data are available on request from the corresponding author, Jason R. Westin (jwestin@mdanderson.org).