1,820
Views
1
CrossRef citations to date
0
Altmetric
Reviews

Utility of measurable residual disease for predicting treatment outcomes with BCR- and BCL2-Targeted therapies in patients with CLL

, , , , , , & show all
Pages 2765-2784 | Received 27 May 2022, Accepted 27 Jun 2022, Published online: 19 Aug 2022

Abstract

Inhibitors targeting B-cell receptor (BCR) signaling pathway proteins and B-cell lymphoma-2 (BCL2) in chronic lymphocytic leukemia (CLL) are recommended in the first-line and relapsed/refractory disease settings. Measurable residual disease (MRD) is an important prognostic tool in patients treated with the BCL2-targeted agent, venetoclax. We explored the relationship between MRD status and progression-free (PFS)/overall survival (OS) in patients with CLL, following treatment with novel BCR- and BCL2-targeted agents. Compared with chemoimmunotherapy, higher rates of undetectable (u)MRD were achieved with BCL2-targeted therapies; achieving uMRD status was associated with longer PFS and OS than MRD-positivity. Continuous treatment with BCR-targeted agents did not achieve uMRD status in many patients, and outcomes were not correlated with uMRD status. Future clinical trials of targeted treatment combinations could be designed to demonstrate uMRD as a treatment objective, and allow a response-driven, personalized strategy to optimize treatment and improve OS outcomes.

Introduction

In trials of chemoimmunotherapy for treatment of patients with chronic lymphocytic leukemia (CLL), achieving undetectable measurable residual disease (uMRD) status at end of treatment (EOT) has been shown to be an independent prognostic marker for longer progression-free (PFS) and overall survival (OS), independent of clinical response status [Citation1–7].

In the phase III CLL8 trial of first-line (1 L) rituximab plus fludarabine and cyclophosphamide (FCR), median PFS was 68.7 months in patients who had uMRD status (<10−4) 2 months after treatment, compared with 40.5 and 15.4 months in patients with intermediate (≥10−4 to <10−2) or high (≥10−2) MRD levels, respectively [Citation6]. Median OS was 48.4 months in patients with high MRD and not reached (NR) in those with uMRD or intermediate-MRD [Citation6]. Combined analysis of data from CLL8 and CLL10 (FCR vs. bendamustine + rituximab [BR]) showed that MRD quantification improved the predictive value of clinical response for PFS outcome [Citation8]. Moreover, later modeling analyses from the German CLL Study Group based on data from >1000 patients treated in the CLL8, CLL10, and CLL11 (obinutuzumab [G] + chlorambucil [Clb] vs. R-Clb vs. Clb) trials demonstrated a statistically significant treatment effect on peripheral blood (PB) uMRD and on PFS, supporting the use of MRD as a surrogate endpoint [Citation9].

However, uMRD rates obtained at EOT from the intent-to-treat (ITT) populations in trials of 1 L chemoimmunotherapy are relatively low (rates for PB and bone marrow [BM] uMRD ranging from 3 to 64% and 1 to 36%, respectively) [Citation6,Citation10–13]. Limited data on MRD status following chemoimmunotherapy are available in the relapsed/refractory (R/R) setting; one study of FCR demonstrated PB and BM uMRD in only 24 and 12%, respectively, of patients from the ITT population with complete response (CR) [Citation14].

Several novel agents targeting key CLL pathogenic pathways are now approved, including the B-cell lymphoma-2 inhibitor (BCL2i) venetoclax (Ven), and B-cell receptor (BCR) signaling pathway inhibitors including Bruton’s tyrosine kinase inhibitors (BTKi’s) ibrutinib (Ibr) and acalabrutinib (Acala), and phosphatidylinositol-3-kinase inhibitors (PI3Ki’s) idelalisib (Idela) and duvelisib [Citation15,Citation16]. BTKi’s and PI3Ki’s are administered continuously until disease progression (PD) [Citation17–20]. This continuous treatment strategy prolongs PFS and OS, but CR rates using the International Workshop on CLL (iwCLL) criteria [Citation21] are low; furthermore, uMRD status is very infrequently attained, thereby reducing the potential for treatment discontinuation and a durable remission off-treatment [Citation22–30]. Therefore, while continuous treatment typically maintains disease control, toxicities, development of resistance, and cost implications of such regimens have led to increasing consideration of fixed-duration therapy options [Citation31].

BCL2i-based therapies result in deep remissions and high uMRD rates, independent of high-risk factors, through fixed-duration treatment [Citation32,Citation33]. Fixed-duration VenG and VenR demonstrated significant efficacy, which was maintained at the 4–5-year follow-ups, with manageable toxicity, in phase III trials in the 1 L (CLL14) and R/R (MURANO) settings, respectively [Citation20,Citation32–35]. Unlike continuous BTKi therapy, fixed-duration treatment with VenG or VenR demonstrates high rates of uMRD in PB and/or BM [Citation32,Citation36–40], with durable remissions off-treatment [Citation32,Citation33]. Thus, the BCL2-targeted treatment strategy is to treat patients for a fixed duration to achieve remission, to monitor their disease during remission, and to plan for re-treatment if/when relapse and progression occur.

Although the association between achieving uMRD status and long-term outcomes has been established for chemoimmunotherapy [Citation5–7], it has been less conclusive with BCL2i-based treatment due to the lesser availability of trial data with targeted agents, and relatively shorter follow-up. We performed a systematic scoping literature review [Citation41] to evaluate and summarize the relationship between MRD response and clinical outcomes (PFS and OS) following treatment with novel BCR- or BCL2-targeted agents in patients with CLL.

Methods

The research question to be investigated was: does uMRD attainment following BCR-targeted or BCL2-targeted treatment correlate with improved PFS and/or OS in patients with CLL?

Data search strategy

Using Medical Subject Heading terms and keywords, the PubMed, Cochrane, and Embase databases were searched for research articles and published abstracts. Registered clinical trials (within clinicaltrials.gov or EudraCT) were also searched in public domains. Keywords used in the search strings are in Supplemental Table 1 and the full list of inclusion/exclusion criteria is shown in Supplemental Table 2.

All trial publications reporting data on MRD and treatment outcomes, using quantitative and/or qualitative assessments, from 1 January 2010 to 31 December 2021 were included, to capture the relevant phase II/III trials for novel targeted agents (US Food and Drug Administration approval of the first novel targeted agent, Ibr, occurred in 2013).

Data screening and extraction

Titles and abstracts of all records (research articles, abstracts, and clinical trial records) were screened in accordance with the inclusion/exclusion criteria by two independent reviewers working in parallel. In cases of discrepancy between the two reviewers, a third reviewer was consulted. Records not meeting the study criteria were excluded. Screening of the full text of ‘included’ publications was then performed by two independent reviewers working in parallel. A third reviewer was used for adjudication in cases of non-alignment between the two reviewers.

The final set of records was then grouped based on the study number/identifier, and key data from these studies were compiled. Details captured from each study were: study population, intervention, doses and treatment duration, MRD assessment method, MRD outcomes, OS, PFS, and key conclusions related to MRD.

Data summary

A descriptive summary of the findings is presented. Unless otherwise specified, all rates of MRD are given for the ITT population (all enrolled/randomized patients); where not provided in the publication, values were re-calculated as a proportion of the ITT population. Generally, for fixed-duration regimens, uMRD rates were reported at EOT; for continuous treatment regimens, best uMRD rates were reported.

Results

Search and study selection

In total, 925 records were obtained from the searches. After excluding duplicates and records not meeting the inclusion criteria, 118 records covering 44 clinical trials were included for analysis (). Summary details of the final included studies are provided in Supplemental Tables 3–5. Details of MRD testing methods, cutoffs for uMRD status and concordance between PB and BM uMRD are provided in the Supplemental Results, Supplemental Figure 1, and Supplemental Table 6.

Figure 1. PRISMA flow chart detailing numbers of included records at each step and reasons for exclusion. aReasons for exclusion: ineligible population (e.g. non-chronic lymphocytic leukemia histology), study designs, interventions or with non-relevant (non-measurable residual disease-related) outcomes, phase I trial, real-word evidence study, non-original data.

Figure 1. PRISMA flow chart detailing numbers of included records at each step and reasons for exclusion. aReasons for exclusion: ineligible population (e.g. non-chronic lymphocytic leukemia histology), study designs, interventions or with non-relevant (non-measurable residual disease-related) outcomes, phase I trial, real-word evidence study, non-original data.

Association between uMRD status achievement with novel targeted therapy and PFS

Details of trials evaluating the association are presented in .

Table 1. Details of trials evaluating PFS and OS association with uMRD status following treatment with novel agents.

BCR-Targeted therapy

Data evaluating association between uMRD attainment and PFS were sparse. The 1 L E1912 trial of continuous Ibr, with R for cycles 2–7, found no significant difference in PFS between patients achieving or not achieving PB uMRD status [Citation43]. However, when MRD was analyzed as a continuous variable on a log10 scale, patients with higher levels of MRD tended to have shorter PFS for each 10-fold increase in MRD level (HR 2.08; 95% CI [confidence interval], 0.84–5.16 at 12 months) [Citation43].

In the R/R HELIOS study of continuous Ibr until PD or unacceptable toxicity, with BR for cycles 1–6, 3-year PFS rates were numerically higher in patients with uMRD vs. those who were MRD-positive (88.6 vs. 60.1% respectively) [Citation44].

BCL2-Targeted therapy

In the 1 L CLL14 study (fixed-duration VenG for 12 months), landmark analysis of PFS from EOT demonstrated a HR of 0.10 for patients with uMRD at EOT (<10−4) vs. those who were MRD-positive; a further landmark analysis showed that patients with uMRD ≤10−5 at EOT had 2-year post-EOT PFS of 93 vs. 37% in patients with MRD levels >10−2 [Citation34,Citation45].

Median PFS was NR in R/R patients with a best response of PB uMRD during treatment with fixed-duration Ven-monotherapy in the VENICE-1 trial compared with 30.5 months in the overall population [Citation49]. Similarly, median PFS was NR in patients with uMRD after a median 16 months of continuous Ven-monotherapy (p = .0019 vs. MRD-positive group [median PFS 21.9 months]) in the M14-032 study in patients with R/R CLL progressing after prior Ibr or Idela therapy [Citation50]. Further, the M13-982 trial of continuous Ven-monotherapy in patients with R/R CLL and chromosome 17p deletion (del(17p)) found that patients who achieved uMRD were less likely to have PD or die compared with patients achieving clinical response but remaining MRD-positive (at median 23 months on therapy) [Citation52].

At the 3-year follow-up of the MURANO trial in patients with R/R disease (fixed-duration VenR for 6 months, followed by Ven-monotherapy up to maximum of 2 years) [Citation36], landmark analysis of PFS from end of combination treatment (EOCT) demonstrated a HR of 0.48 for patients with uMRD vs. intermediate-MRD-positive status, 0.15 for uMRD vs. high-MRD-positive and 0.24 for intermediate-MRD-positive vs. high-MRD-positive status. This was sustained at the 4-year follow-up [Citation53]. At the 5-year follow-up (3 years post-EOT), 32/83 patients (39%) with uMRD at EOT remained in ongoing uMRD status; baseline presence of high-risk prognostic factors was associated with increased risk of MRD conversion and PD [Citation35].

Combination BCR-/BCL2-targeted therapy

At 1 year of follow-up in the 1 L GLOW trial, PFS was similar for patients with/without uMRD status in the fixed-duration Ven-Ibr arm, while patients treated with G-Clb who achieved uMRD status had longer PFS than those with detectable MRD [Citation54].

Association between uMRD status and OS

Findings are available in .

BCR-targeted therapy

No difference in OS in relation to MRD status was observed in patients who responded to Ibr-BR treatment in the HELIOS study on multivariate analysis (median 34.8 months on-study) [Citation44].

BCL2-targeted therapy

The fixed-duration regimen studies, CLL14 and MURANO, indicated that achieving uMRD status after VenG or VenR, respectively, was linked to improved OS. A CLL14 post-hoc analysis found longer OS in patients with uMRD at EOT (medians NR, median follow-up 39.6 months) [Citation46]. Similarly, in MURANO, achieving uMRD status at EOT with VenR was associated with a trend toward improved OS (OS 3 years post-EOT 95 vs. 85% in patients with uMRD vs. MRD-positive status at EOT, respectively) [Citation35].

Independence of uMRD from clinical response as a prognostic marker

BCR-targeted therapy

Findings from the 1 L E1912 trial showed that MRD-positive patients without CR at 12 months of treatment had significantly worse PFS compared with patients with CR or uMRD status (HR 3.73; 95% CI, 1.14–12.27) [Citation43]. Further, patients with uMRD status had similar PFS regardless of CR status, although patient numbers were small.

BCL2-targeted therapy

Landmark analysis of PFS according to uMRD status at EOT in CLL14 showed that the improved PFS with uMRD vs. MRD-positive status (HR 0.10; 95% CI, 0.06–0.15) was seen regardless of clinical response (in a combined analysis of patients treated with 1 L VenG and G-Clb) [Citation46].

Similarly, at the 3-year follow-up of the MURANO trial [Citation36], improved PFS was observed in R/R patients who achieved uMRD at EOCT, regardless of clinical response or treatment arm at EOCT (). Landmark analysis showed that VenR-treated patients achieving partial response (PR) and uMRD at EOCT had similar PFS to those achieving CR and uMRD (HR 0.71; 95% CI, 0.24–2.14). Patients with PR and detectable MRD had inferior PFS after EOCT vs. those with PR and uMRD in Kaplan–Meier analysis [Citation36]. At 4-years’ follow-up, MRD status did not seem to affect PFS in patients achieving CR/CR with incomplete hematologic recovery [Citation53].

Rates of uMRD achieved with targeted therapies

BCR-targeted therapy

In the 1L setting (), the lowest rates of PB and BM uMRD were seen in trials of continuous Ibr-monotherapy (e.g. 1% PB uMRD after 1 year, GELLC-7 [Citation55]; 6% after 4 years, NCT01500733) [Citation42]. Rates increased with addition of a CD20 monoclonal antibody (mAb), with a uMRD rate of 30% after ∼2.5 years in the iLLUMINATE trial (continuous Ibr with G for cycles 1–6) [Citation30]. BM uMRD rates following 1 L treatment were highest in the NCT02007044 trial of IbrR (25%, although numbers were small [n = 12]) [Citation56], and the iLLUMINATE trial of IbrG (20%) [Citation30].

Figure 2. Rates of uMRD during continuous BCR-targeted therapy in the 1L setting (A), for R/R disease (B) and in trials with an MRD- or response-guided component (C) (ITT populations). 1L: first-line; B: bendamustine; BCR: B-cell receptor; BM: bone marrow; EOCT: end of combination treatment; FU: follow-up; G: obinutuzumab; Ibr: ibrutinib; Idela: idelalisib; iFCG,: ibrutinib plus fludarabine, cyclophosphamide, and G; ITT: intent-to-treat; mono: monotherapy; MRD: measurable residual disease; mo: months; NR: not reported; PB: peripheral blood; R: rituximab; R/R: relapsed/refractory; Tafa: tafasitamab; Ubli: ublituximab; uMRD: undetectable MRD. Details of treatment regimens and MRD analysis methodology are provided in Tables S3–S5. a92% of patients also received four cycles of standard-dose FCG.

Figure 2. Rates of uMRD during continuous BCR-targeted therapy in the 1L setting (A), for R/R disease (B) and in trials with an MRD- or response-guided component (C) (ITT populations). 1L: first-line; B: bendamustine; BCR: B-cell receptor; BM: bone marrow; EOCT: end of combination treatment; FU: follow-up; G: obinutuzumab; Ibr: ibrutinib; Idela: idelalisib; iFCG,: ibrutinib plus fludarabine, cyclophosphamide, and G; ITT: intent-to-treat; mono: monotherapy; MRD: measurable residual disease; mo: months; NR: not reported; PB: peripheral blood; R: rituximab; R/R: relapsed/refractory; Tafa: tafasitamab; Ubli: ublituximab; uMRD: undetectable MRD. Details of treatment regimens and MRD analysis methodology are provided in Tables S3–S5. a92% of patients also received four cycles of standard-dose FCG.

In the R/R setting (), rates of PB uMRD ranged from 7% with ublituximab (Ubli)-Ibr after 6 months of follow up to 46% after 42 months of follow-up (GENUINE) [Citation57].

In 1 L trials of Ibr that included a response- and/or MRD-guided maintenance component, a substantial number of patients achieved or maintained uMRD following maintenance ().

BCL2-targeted therapy

Trials of BCL2-targeted therapies mainly evaluated fixed-duration regimens. PB uMRD rates achieved at EOT in the 1 L setting ranged between 38% with BR-VenR (3 cycles BR, followed by 12 Ven-containing cycles; NCT03609593) [Citation58] and 89% with VenG (CLL13) [Citation59] (). Rates were generally lower in the R/R setting, with the highest rate achieved with VenR in the MURANO trial (64%) [Citation32]. BM uMRD rates achieved at EOT in 1 L trials ranged from 35% with BR-VenR (NCT03609593) [Citation58] to 73% with VenG (CLL13) [Citation59]. One trial of BCL2-targeted therapy in the R/R setting reported a BM uMRD rate of 50% with atezolizumab-VenG (NCT02846623) [Citation60]. Two trials evaluated continuous treatment with Ven-monotherapy in the R/R setting, demonstrating PB uMRD rates of 22–30% (M14-032 and M13-982); one additional trial had a uMRD rate of 46% with the CD19 mAb tafasitamab (Tafa) plus Ven (COSMOS cohort B) () [Citation61].

Figure 3. Rates of uMRD at EOTa following fixed-duration BCL2-targeted therapy (A),b during continuous BCL2-targeted therapy (B), and in trials with an MRD- or response-guided component (C)c (ITT populations). 1L: first-line; Atezo: atezolizumab; B: bendamustine; BCL2: B-cell lymphoma-2; BM: bone marrow; EOT: end of treatment; FU: follow-up; G: obinutuzumab; Ibr: ibrutinib; ITT: intent-to-treat; mo: months; mono: monotherapy; MRD: measurable residual disease; NR: not reported; PB: peripheral blood; R: rituximab; R/R: relapsed/refractory; Tafa: tafasitamab; uMRD: undetectable MRD; Ven: venetoclax; wks: weeks. Details of treatment regimens and MRD analysis methodology are provided in Tables S3–S5. aValues within 3 mo of EOT are included. bMURANO trial: n represents the number of patients who completed the full 2 years of Ven treatment; HOVON 139/GIVE trial: n represents the number of patients randomized to Arm A: fixed-duration Ven maintenance for 12 cycles following induction; values reported are at end of maintenance. cHOVON 139/GIVE trial: Only two patients randomized to the MRD-guided arm actually received Ven due to MRD-positivity.

Figure 3. Rates of uMRD at EOTa following fixed-duration BCL2-targeted therapy (A),b during continuous BCL2-targeted therapy (B), and in trials with an MRD- or response-guided component (C)c (ITT populations). 1L: first-line; Atezo: atezolizumab; B: bendamustine; BCL2: B-cell lymphoma-2; BM: bone marrow; EOT: end of treatment; FU: follow-up; G: obinutuzumab; Ibr: ibrutinib; ITT: intent-to-treat; mo: months; mono: monotherapy; MRD: measurable residual disease; NR: not reported; PB: peripheral blood; R: rituximab; R/R: relapsed/refractory; Tafa: tafasitamab; uMRD: undetectable MRD; Ven: venetoclax; wks: weeks. Details of treatment regimens and MRD analysis methodology are provided in Tables S3–S5. aValues within 3 mo of EOT are included. bMURANO trial: n represents the number of patients who completed the full 2 years of Ven treatment; HOVON 139/GIVE trial: n represents the number of patients randomized to Arm A: fixed-duration Ven maintenance for 12 cycles following induction; values reported are at end of maintenance. cHOVON 139/GIVE trial: Only two patients randomized to the MRD-guided arm actually received Ven due to MRD-positivity.

In the HOVON 139/GIVE trial of 1 L VenG, following treatment cycle 14, patients with a clinical response were randomized to fixed-duration treatment with 12 cycles of Ven-monotherapy or MRD-guided Ven (in which MRD-positive patients at cycle 14 received 12 cycles of Ven-monotherapy and patients with uMRD status discontinued). Following 12 months of treatment or observation in the MRD-guided group, 57% of patients had maintained or achieved uMRD status (). The IMPROVE trial of Ven-monotherapy in R/R disease showed that 84% of the 19 patients (of 38; 50%) who did not achieve uMRD status following 12 cycles of Ven-monotherapy achieved uMRD status following 12 months’ Ven-Ibr maintenance ().

Combination BCR-/BCL2-targeted therapy

One 1 L trial of fixed-duration combination BCL2 and BCR-targeted therapy demonstrated PB and BM uMRD rates of 88 and 78%, respectively, with Ven-IbrG (CLL13) () [Citation59].

Figure 4. Rates of uMRD during combination BCR- and BCL2-targeted therapy of fixed duration (A), as continuous treatment (B) and in trials with an MRD- or response-guided component (C and D) (ITT populations). 1L: first-line; BCL2: B-cell lymphoma-2; BCR: B-cell receptor; BM: bone marrow; EOT: end of treatment; FU: follow-up; G: obinutuzumab; Ibr: ibrutinib; ITT: intent-to-treat; mo: months; MRD: measurable residual disease; NR: not reported; PB: peripheral blood; R/R: relapsed/refractory; uMRD: undetectable MRD; Ven: venetoclax. Details of treatment regimens and MRD analysis methodology are provided in Tables S3–S5.

In the R/R setting, the CLARITY trial showed BM uMRD rates of 35% after 1 year and 44% after 2 years of continuous Ven-Ibr treatment () [Citation62].

In the 1 L CAPTIVATE trial MRD cohort, similar PB uMRD rates were observed in patients following 12 months’ maintenance with placebo or Ibr-monotherapy (84 and 77%, respectively) following uMRD achievement with 12 months’ combination therapy with Ven-Ibr () [Citation63]. Similarly, 12 months after randomization to observation or Ibr-monotherapy following achievement of CR plus uMRD at 15 cycles of Ven-Ibr for R/R disease, similar rates of BM uMRD were observed (71% and 75%, respectively) in the VISION-HO141 trial () [Citation64].

uMRD rates in patients with high-risk genetic features

BCR-targeted therapy

Most BCR-targeted therapy trials showed similar rates of uMRD in patients with high-risk genetics compared with the overall population (). Multivariate analysis from the E1912 trial (IbrR) found that having mutated IGHV was associated with achieving uMRD status [Citation43].

Table 2. Rates of uMRD in patients with high-risk genetics compared with the overall population.

BCL2-targeted therapy

Similar to findings with BCR-targeted therapy, the rates of uMRD achieved by BCL2-targeted therapies were generally comparable between patients with high-risk genetics and the overall population (). In the 1 L setting, this was observed in the CLL14 and CLL2-BAG trials [Citation33,Citation46,Citation76]. In the R/R setting, the MURANO trial (VenR) found that uMRD status at EOT was not associated with four major cytogenetic alterations (del(17p), chromosome 11q deletion, Trisomy 12, or chromosome 13q deletion) [Citation36,Citation53]. High and low karyotypic complexity (≥3 or ≥5 structural and/or numerical chromosomal aberrations, respectively [Citation84]) correlated with MRD-positive status at EOT; 36% of patients without complex karyotype (CK) were MRD-positive, compared with 42% and 50% of patients with low and high CK, respectively [Citation59,Citation77,Citation85]. Further, patients treated with VenR who had TP53, NOTCH1, XPO1, or BRAF mutations had numerically lower rates of uMRD.

Combination BCR-/BCL2-targeted therapy

Similar patterns were found when BCR and BCL2-targeted therapies were given in combination, with the exception of the AVO (Acala-VenG) trial in 1 L [Citation82], which reported lower rates of uMRD in patients with high-risk genetics compared with the overall study population. A univariate analysis using data from patients treated with Ven-IbrG in the OSU-14266 trial found no association between high-risk genetic markers including unmutated IGHV, presence of del(17p), or CK and achievement of uMRD status [Citation86].

MRD responses below 10−4 following treatment with novel oral targeted agents

BCR-targeted therapy

At EOT in the ICLL-07 trial (following 9 months of IbrG and 6 months of Ibr plus FCG therapy [iFCG; in 92% of patients]) [Citation66], 67% of patients had BM uMRD at a cutoff <10−6 (79% <10−4). In a phase II trial in 45 patients (NCT02629809) [Citation87], BM uMRD <10−5 assessed via next-generation sequencing (NGS) was achieved after three cycles of iFCG by 56% of patients and by 42% at <10−6 (87% <10−4 by flow cytometry), increasing to 71 and 47%, respectively (89% <10−4) after a further three cycles of IbrG, and 78 and 51%, respectively, after a further six cycles of Ibr-monotherapy or IbrG (91% <10−4) [Citation87,Citation88].

BCL2-targeted therapy

In CLL14 [Citation45], at 3 months post-EOT, 42% of VenG-treated patients had PB uMRD <10−6 (75% <10−4).

Combination BCR-/BCL2-targeted therapy

The CLARITY study reported that, after 12 months of Ven-Ibr in R/R patients, 29% of patients had uMRD <10−5 (35% <10−4) [Citation89].

uMRD response duration

BCR-targeted therapy

Of 61 patients who began Ibr maintenance with BM uMRD status following iFCR in the NCT02251548 trial, 13 (21%) had recurrent BM MRD positivity; median time to MRD recurrence NR [Citation90]. In a phase II trial of Ubli plus umbralisib (U2)-Ibr, of 16 patients who entered treatment-free observation (15 due to achievement of uMRD), no progression was observed at median 242 days off treatment [Citation91].

BCL2-targeted therapy

In CLL14, uMRD was sustained in many patients throughout treatment up to month 18 of follow-up [Citation45]; PB uMRD was achieved by 75% of VenG-treated patients 3 months post-EOT, 81% 12 months post-EOT, 47% 18 months post-EOT, and 27% 30 months post-EOT [Citation92]. At 12 months post-EOT, the HR for MRD conversion was 0.19 (95% CI, 0.12–0.30). At a median follow-up of 39.6 months, 10 of 90 patients with uMRD <10−6 at EOT converted to MRD positivity, including one patient with PD; 18-month conversion-free survival was 88%.

In MURANO [Citation32,Citation53], 62% of patients had uMRD at EOCT (after 6 months’ VenR) and 48% had uMRD at EOT (after 2 years’ Ven). Of the 83 patients who completed 2 years of Ven and had uMRD at EOT, 70% had uMRD status at a median 9.9 months post-EOT, and 39% had ongoing uMRD status at 3 years post-EOT [Citation32, Citation35,Citation36, Citation53].

Combination BCR-/BCL2-targeted therapies

In GLOW [Citation78], 55% of patients receiving 1 L fixed-duration Ven-Ibr achieved uMRD status 3 months post-EOT, of whom 85% maintained uMRD status to 12 months post-EOT [Citation54,Citation78]. In the 1 L cohort of the NCT02756897 trial [Citation93], after a median follow-up of 12.4 months, 8/53 (15%) patients with uMRD after 24 cycles of Ven-Ibr had MRD recurrence, five of whom had first achieved uMRD status at completion of 24 cycles. In a phase II trial of Ven-Ibr from the MD Anderson Cancer Center, at a median follow-up of 29 months, 29/45 (64%) patients have achieved uMRD, with two patients subsequently progressing [Citation94]. Of the patients receiving U2-Ven in the NCT03801525 trial, 45% (18/40) had PB uMRD at EOT; 18% (7/40) had uMRD 6 months post-EOT, and 10% (4/40) had uMRD 12 months post-EOT [Citation95,Citation96].

Discussion

We explored whether attaining uMRD status following treatment with BCR- or BCL2-targeted therapies, either as fixed-duration or continuous treatment regimens, including regimens with an MRD-guided treatment component (for 1 L treatment or R/R disease), correlated with longer PFS and OS in patients with CLL, compared with an MRD-positive status. BCR-targeted therapies resulted in low rates of uMRD attainment; however, achievement of uMRD was associated with improved PFS vs. MRD-positivity. BCL2-targeted agents generally achieved deep, durable responses, with high rates of uMRD; achievement of uMRD was associated with prolonged PFS, independent of clinical response status, and prolonged OS.

BCR-targeted therapy

Achievement of PB uMRD <10−4 was infrequent in patients treated with Ibr in 1 L (rate with Ibr-monotherapy, 1–6%), although rates were increased with the addition of a CD20 mAb, in particular G (30% after 2.5 years with IbrG in iLLUMINATE) [Citation30], and prolonged duration of treatment. BM uMRD rates showed a similar pattern. In patients with R/R disease, the highest rate of BM uMRD was achieved by Ubli-Ibr in the GENUINE trial (46% after 42 months’ follow-up) [Citation72]. BM uMRD data were sparse in R/R trials.

Deep responses were achievable in a proportion of patients who received Ibr plus chemotherapy, with uMRD <10−6 rates of 42–67% reported in patients who received iFCG in the ICLL-07 and NCT02629809 trials [Citation66,Citation88]. Rates of uMRD achieved were generally comparable between patients with high-risk cytogenetics and the overall trial populations, although no data on depth of response were available in high-risk patients.

Data on association between uMRD and PFS with BTKi’s were limited, and likely impacted by the low rates of uMRD attainment. One trial evaluated PFS outcomes in patients with uMRD at <10−4 and MRD-positive patients following 1 L IbrR (E1912), and found no significant difference between the groups at any timepoint, although PFS was longer in patients with MRD < vs. >10−1 [Citation43]. However, in the R/R HELIOS trial, after a median of 35 months on therapy, Ibr-BR was shown to improve PFS over placebo-BR in patients with uMRD, intermediate-MRD-positive status, and high-MRD-positive status. Multivariate analysis from HELIOS showed no difference in OS among these response categories.

BCL2-targeted therapies

High PB uMRD rates were seen following fixed-duration BCL2-targeted therapy with Ven-containing regimens in the 1 L and R/R settings. Trials of VenG and VenR demonstrated rates of 72–89% [Citation33,Citation59,Citation74], with the exception of the NCT03609593 trial [Citation58], which had a lower PB uMRD rate of 38% following BR-VenR treatment, perhaps due to compromised Ven delivery owing to myelosuppression with the addition of bendamustine. BM uMRD rates followed a similar pattern. Fewer data were available in the R/R setting, with the highest PB uMRD rates observed with VenR in the MURANO trial [Citation32]. Rates of PB uMRD in patients treated with continuous Ven-monotherapy were lower, ranging from 22 to 30%, although a small trial of Tafa-Ven demonstrated a uMRD rate of 46% after 15 months [Citation61]. There was a lack of data on MRD-guided maintenance following Ven-based treatment; one small study in R/R CLL found that 16 of 19 patients who did not achieve uMRD with VenG went on to attain uMRD status following 12 months’ Ibr maintenance [Citation97].

Similar to BCR-targeted therapies, most trials reported comparable uMRD rates in patients with high-risk genetics vs. the overall study population. However, earlier reemergence of MRD and shorter PFS have been observed for patients with high-risk biological features treated with fixed-duration Ven-based regimens, indicating more rapid kinetics of disease regrowth [Citation92]. Notably, genomic complexity has not been associated with significantly inferior outcomes in 1 L Ven-based treatment, but more so in the R/R setting [Citation53,Citation98]. Longer follow-up is needed, as divergence in outcomes based on presence/absence of genomic complexity may subsequently emerge, but this suggests that early Ven treatment may overcome this risk feature to some degree.

uMRD status achieved with fixed-duration Ven-based regimens was typically sustainable for more than 12 months post-EOT in the majority of patients, with prolonged PFS even after patients stopped therapy. Data from the CLL14 and MURANO trials indicate that uMRD achievement following fixed-duration Ven-based therapies is associated with prolonged PFS, and potentially OS; however, it is still too early to accurately assess the impact on OS, given the overall good prognosis, few deaths, and availability of effective salvage regimens for patients receiving targeted therapies. Therefore, longer follow-up of these clinical trials is needed. This aligns with findings from a quantitative meta-analysis of 11 chemotherapy and chemoimmunotherapy studies (median follow-up 28 months–12.8 years) showing significant association between uMRD status and OS, which was not maintained when considering only patients who achieved CR [Citation4]. As clinical trials with extended follow-up indicate that patients achieving a deep response with uMRD have better long-term survival than those with persistent MRD [Citation39], therapy directed at eradication of detectable MRD may become a desirable goal in clinical practice.

Combination BCR- and BCL2-targeted therapies

Data on association between uMRD attainment and PFS in patients treated with combination BCR- and BCL2-targeted therapies are limited to date. Early data from the 1 L GLOW trial of Ven-Ibr found similar PFS of 90% at 12-months post-EOT; although in the chemoimmunotherapy comparator arm, it was seen that MRD-positive patients relapsed more quickly than those with uMRD [Citation54].

The CLL13 trial demonstrated high PB and BM uMRD rates following 1 L Ven-IbrG of 88 and 78%, respectively [Citation59]. The highest rates of PB and BM uMRD seen in the R/R setting with a continuous treatment regimen were with Ven-Ibr in the CLARITY trial [Citation62], although rates were lower than those achieved with fixed-duration VenR in MURANO [Citation32].

MRD assessment methodology

Flow cytometry (mainly 4-color [sensitivity 10−4] [Citation99] and 8-color [sensitivity 10−5] [Citation100]), was the most commonly used method for MRD detection for clinical trials included in this analysis. This fits with the recent review by Al Sawaf et al. [Citation47] which highlights harmonization efforts by the European Research Initiative on CLL [Citation101] for flow cytometry MRD assessment across trials, and compares with polymerase chain reaction and NGS testing. While deeper response levels (MRD levels <10−5 or lower) are informative for greater depth of remissions, allowing serial tracking of response kinetics (and re-growth), currently the <10−4 threshold should be maintained, as it is most robustly correlated with long-term clinical outcomes from the chemoimmunotherapy era. However, future investigations of more potent novel targeted treatment combinations in clinical trials should explore the potential impact of deeper responses. Furthermore, to be relevant for standard-of-care treatment, and particularly in relation to MRD-directed therapy, future clinical investigations will need to utilize MRD methodology that meets the regulatory standards. Although few studies assessed concordance levels between MRD assessment based on PB and BM, reported concordance levels were high. This suggests that assessment of MRD in PB, which is more convenient than BM sampling, is highly reliable and also appropriate for serial clinical trial assessments.

Limitations

Due to variations in study protocols/methodology across the final set of studies considered in this review, conclusive evidence on the research question could not be established. Risk of bias is inherent with this type of analysis; however, to reduce this risk, first- and second-pass screening was carried out by two independent reviewers, and results were subject to adjudication and reconciliation at each stage. However, publication bias could not be excluded. Immortal-time bias may also be present, particularly for studies that did not clearly define the starting point for time-to-event endpoint calculations.

Clinical perspective

Approved fixed-duration targeted therapy for CLL currently consists of Ven-based regimens. Trials of MRD-directed therapy are lacking generally; no such trials have been conducted for chemoimmunotherapy, and data are very limited for Ven-based treatment. Yet, as outlined in this article, an MRD-guided strategy may provide additional benefits to fixed-duration or continuous treatment regimens in terms of improved efficacy and quality of life, or reduced toxicity and exposure. Clinical trials are needed to address this question. The use of Ven-based consolidation strategies is also relevant, with the aim of deepening remission by converting continuous treatment to fixed-duration treatment.

A substantial body of evidence correlates uMRD status with longer PFS and OS following fixed-duration Ven-based treatment, irrespective of whether Ven is used as monotherapy, or in combination with a CD20 mAb. It remains to be seen whether Ven in combination with other targeted agents (BTKi’s or PI3Ki’s) achieves long-term survival.

Targeted therapy with BCR signaling pathway small molecule inhibitors (BTKi’s and PI3Ki’s) is administered continuously without expectation of uMRD in a notable proportion of patients. Thus, if a patient on such treatment achieves uMRD status, an option may be to discontinue treatment.

The question remains as to whether MRD testing should be recommended outside of clinical trials. Our review does not address the limitations of using MRD to assess clinical outcome in the community practice setting, nor does it address the implications of MRD assessment for patients of different genetic high-risk subgroups. However, uMRD achievement in the context of fixed-duration Ven-based treatment has stronger prognostic value than iwCLL response, since it is much more widely evaluated and independently validated (with iwCLL response being largely based on consensus regarding arbitrary cutoffs rather than disease biology). Already, MRD status has current clinical implications where prognostication is desired e.g. at EOT, and therefore wider adoption of MRD assessment warrants consideration.

Conclusions

MRD status is an important prognostic tool in patients with CLL receiving fixed-duration therapy. BCL2-targeted therapy achieves uMRD status in a high proportion of patients with fixed-duration treatment, and uMRD status is correlated with longer PFS and OS than MRD-positive status, as with chemoimmunotherapy. Compared with chemoimmunotherapy, higher rates of uMRD are achieved with Ven-based targeted therapies. However, as data are limited, we can only determine that the association of uMRD status and PFS is independent of CR in Ven-based studies.

BCR pathway inhibitor-based treatment (BTKi’s and PI3Ki’s) is continuous and does not achieve uMRD status for the vast majority of patients. Thus, consideration of the association of uMRD status with outcome is less relevant for these agents. Combination targeted trials (BTKi’s plus BCL2i) have often used fixed‐duration treatment and have achieved high uMRD rates, but follow-up is too short to allow firm correlations to be drawn. Future clinical trials of novel targeted treatments should explore MRD-guided therapy approaches and include longer follow-up to assess the impact of uMRD with targeted treatments on OS.

Author contributions

All authors were involved in interpreting the data and writing the manuscript. All authors approved the final version for submission.

Supplemental material

GLAL-2022-0520-File002.docx

Download MS Word (344.5 KB)

Acknowledgments

Venetoclax is being developed in a collaboration between Genentech, Inc. and AbbVie. Third-party medical writing assistance, under the direction of the authors, was provided by Lynda McEvoy, PhD, and Rachel Dobb, PhD, of Ashfield MedComms, an Ashfield Health company, and was funded by F. Hoffmann-La Roche Ltd.

Disclosure statement

WGW is employed by The University of Texas MD Anderson Cancer Center; and has received research funding from AbbVie and Genentech, Inc; and travel, accommodations, and expenses from AbbVie and Genentech, Inc. TJK owns stock in and patents/royalties from Oncternal Therapeutics Inc; has received research funding from Pharmacyclics, AbbVie, Breast Cancer Research Foundation, MD Anderson Cancer Center, Oncternal Therapeutics Inc, Specialized Center of Research (SCOR) – The Leukemia and Lymphoma Society (LLS), California Institute for Regenerative Medicine (CIRM), and the National Cancer Institute/NIH; and honoraria from Pharmacyclics, AbbVie, Genentech, Inc., F. Hoffmann-La Roche Ltd, Janssen, Gilead, National Cancer Institute/NIH, and Celgene. Cirmtuzumab was developed by TJK in the TJK laboratory and licensed by the University of California to Oncternal Therapeutics, Inc., which provided stock options and research funding to the TJK laboratory. OA-S has received honoraria from F. Hoffmann-La Roche Ltd, Janssen, Gilead, AbbVie, AstraZeneca, BeiGene, and Adaptive; has participated in advisory boards and received personal fees from F. Hoffmann-La Roche Ltd, Janssen, Gilead, AbbVie, and AstraZeneca; and has received research funding from BeiGene, F. Hoffmann-La Roche Ltd, Janssen, and AbbVie. BC is employed by and owns equity in AbbVie. JMLB is employed by Genentech, Inc./F. Hoffmann-La Roche Ltd and owns equity in F. Hoffmann-La Roche Ltd. YM is employed by Genentech, Inc. and owns equity in F. Hoffmann-La Roche Ltd. YJ is employed by Genentech, Inc./F. Hoffmann-La Roche Ltd and owns equity in F. Hoffmann-La Roche Ltd. JFS has received consultancy fees from Celgene and F. Hoffmann-La Roche Ltd; research funding from AbbVie, Celgene, Janssen, and F. Hoffmann-La Roche Ltd; has participated in speakers bureau for AbbVie, Celgene, and F. Hoffmann-La Roche Ltd; has membership of an entity’s Board of Directors or has participated in advisory committees for, and received honoraria from, AbbVie, AstraZeneca, BMS, Gilead, Janssen, Mei Pharma, Morphosys, F. Hoffmann-La Roche Ltd, Sunesis, and Takeda.

Data availability statement

The authors confirm that the data supporting the findings of this study are available within the article and its supplementary materials.

Additional information

Funding

This comprehensive literature review was supported by F. Hoffmann-La Roche Ltd.

References

  • Burger JA. Treatment of chronic lymphocytic leukemia. N Engl J Med. 2020;383(5):460–473.
  • Thompson PA, Peterson CB, Strati P, et al. Serial minimal residual disease (MRD) monitoring during first-line FCR treatment for CLL may direct individualized therapeutic strategies. Leukemia. 2018;32(11):2388–2398.
  • Del Giudice I, Raponi S, Della Starza I, et al. Minimal residual disease in chronic lymphocytic leukemia: a new goal? Front Oncol. 2019;9:689.
  • Molica S, Giannarelli D, Montserrat E. Minimal residual disease and survival outcomes in patients with chronic lymphocytic leukemia: a systematic review and meta-analysis. Clin Lymphoma Myeloma Leuk. 2019;19(7):423–430.
  • Fischer K, Cramer P, Busch R, et al. Bendamustine in combination with rituximab for previously untreated patients with chronic lymphocytic leukemia: a multicenter phase II trial of the German chronic lymphocytic leukemia study group. J Clin Oncol. 2012;30(26):3209–3216.
  • Bottcher S, Ritgen M, Fischer K, et al. Minimal residual disease quantification is an independent predictor of progression-free and overall survival in chronic lymphocytic leukemia: a multivariate analysis from the randomized GCLLSG CLL8 trial. J Clin Oncol. 2012;30(9):980–988.
  • Strati P, Keating MJ, O'Brien SM, et al. Eradication of bone marrow minimal residual disease may prompt early treatment discontinuation in CLL. Blood. 2014;123(24):3727–3732.
  • Kovacs G, Robrecht S, Fink AM, et al. Minimal residual disease assessment improves prediction of outcome in patients with chronic lymphocytic leukemia (CLL) who achieve partial response: comprehensive analysis of two phase III studies of the German CLL study group. J Clin Oncol. 2016;34(31):3758–3765.
  • Dimier N, Delmar P, Ward C, et al. A model for predicting effect of treatment on progression-free survival using MRD as a surrogate end point in CLL. Blood. 2018;131(9):955–962.
  • Hallek M, Fischer K, Fingerle-Rowson G, et al. Addition of rituximab to fludarabine and cyclophosphamide in patients with chronic lymphocytic leukaemia: a randomised, open-label, phase 3 trial. Lancet. 2010;376(9747):1164–1174.
  • Eichhorst B, Fink AM, Bahlo J, German CLL Study Group (GCLLSG), et al. First-line chemoimmunotherapy with bendamustine and rituximab versus fludarabine, cyclophosphamide, and rituximab in patients with advanced chronic lymphocytic leukaemia (CLL10): an international, open-label, randomised, phase 3, non-inferiority trial. Lancet Oncol. 2016;17(7):928–942.
  • Goede V, Fischer K, Busch R, et al. Obinutuzumab plus chlorambucil in patients with CLL and coexisting conditions. N Engl J Med. 2014;370(12):1101–1110.
  • Bosch F, Cantin G, Cortelezzi A, et al. Obinutuzumab plus fludarabine and cyclophosphamide in previously untreated, fit patients with chronic lymphocytic leukemia: a subgroup analysis of the GREEN study. Leukemia. 2020;34(2):441–450.
  • Robak T, Dmoszynska A, Solal-Céligny P, et al. Rituximab plus fludarabine and cyclophosphamide prolongs progression-free survival compared with fludarabine and cyclophosphamide alone in previously treated chronic lymphocytic leukemia. J Clin Oncol. 2010;28(10):1756–1765.
  • Burger JA, O'Brien S. Evolution of CLL treatment - from chemoimmunotherapy to targeted and individualized therapy. Nat Rev Clin Oncol. 2018;15(8):510–527.
  • Eichhorst B, Robak T, Montserrat E, ESMO Guidelines Committee. Electronic address: [email protected], et al. Chronic lymphocytic leukaemia: ESMO clinical practice guidelines for diagnosis, treatment and follow-up. Ann Oncol. 2021;32(1):23–33.
  • FDA. Calquence. Highlights of prescribing information; 2021 [cited 2021 Jun 17]. Available from: https://www.azpicentral.com/calquence/calquence.pdf#page=1.
  • FDA. Copiktra. Highlights of prescribing information; [cited 2021 Jun 17]. Available from: https://copiktra.com/pdf/verastem/COPIKTRA-PI-072019.pdf.
  • FDA. Imbruvica. Highlights of prescribing information; 2021 [cited 2021 Jun 17]. Available from: https://imbruvica.com/files/prescribing-information.pdf.
  • FDA. Zydelig. Highlights of prescribing information; 2021 [cited 2021 Jun 17]. Available from: https://www.gilead.com/∼/media/Files/pdfs/medicines/oncology/zydelig/zydelig_pi.pdf.
  • Hallek M, Cheson BD, Catovsky D, International Workshop on Chronic Lymphocytic Leukemia, et al. Guidelines for the diagnosis and treatment of chronic lymphocytic leukemia: a report from the international workshop on chronic lymphocytic leukemia updating the National Cancer Institute-Working group 1996 guidelines. Blood. 2008;111(12):5446–5456.
  • Munir T, Rawstron A, Brock K, et al. Ibrutinib plus venetoclax in relapsed/refractory CLL: results of the bloodwise TAP clarity study. Br J Haematol. 2019;185, 25 (conference paper).
  • Byrd JC, Hillmen P, O’Brien S, et al. Long-term follow-up of the RESONATE phase 3 trial of ibrutinib vs ofatumumab. Blood. 2019;133(19):2031–2042.
  • Burger JA, Tedeschi A, Barr PM, et al. Ibrutinib as initial therapy for patients with chronic lymphocytic leukemia. N Engl J Med. 2015;373(25):2425–2437.
  • Woyach JA, Ruppert AS, Heerema NA, et al. Ibrutinib regimens versus chemoimmunotherapy in older patients with untreated CLL. N Engl J Med. 2018;379(26):2517–2528.
  • Shanafelt TD, Wang XV, Kay NE, et al. Ibrutinib–rituximab or chemoimmunotherapy for chronic lymphocytic leukemia. N Engl J Med. 2019;381(5):432–443.
  • Lampson BL, Tyekucheva S, Crombie JL, et al. Preliminary safety and efficacy results from a phase 2 study of acalabrutinib, venetoclax and obinutuzumab in patients with previously untreated chronic lymphocytic leukemia (CLL). Blood. 2019;134(Supplement_1):e470–e479.
  • Davids MS, Fisher DC, Tyekucheva S, et al. A phase 1b/2 study of duvelisib in combination with FCR (DFCR) for frontline therapy for younger CLL patients. Leukemia. 2021;35(4):1064–1072.
  • Sharman JP, Coutre SE, Furman RR, et al. Final results of a randomized, phase III study of rituximab with or without idelalisib followed by open-label idelalisib in patients with relapsed chronic lymphocytic leukemia. J Clin Oncol. 2019;37(16):1391–1402.
  • Moreno C, Greil R, Demirkan F, et al. Ibrutinib plus obinutuzumab versus chlorambucil plus obinutuzumab in first-line treatment of chronic lymphocytic leukaemia (iLLUMINATE): a multicentre, randomised, open-label, phase 3 trial. Lancet Oncol. 2019;20(1):43–56.
  • Molica S. The evolving role of time-limited targeted therapy in chronic lymphocytic leukemia. Expert Rev Anticancer Ther. 2020;20(12):1015–1019.
  • Seymour JF, Kipps TJ, Eichhorst B, et al. Venetoclax-rituximab in relapsed or refractory chronic lymphocytic leukemia. N Engl J Med. 2018;378(12):1107–1120.
  • Fischer K, Al-Sawaf O, Bahlo J, et al. Venetoclax and obinutuzumab in patients with CLL and coexisting conditions. N Engl J Med. 2019;380(23):2225–2236.
  • Al-Sawaf O, Zhang C, Robrecht S, et al. Venetoclax-obinutuzumab for previously untreated chronic lymphocytic leukemia: 4-year follow-up analysis of the randomized CLL14 study. Abstract Book for the 26th Congress of the European Hematology Association Virtual Edition, 2021, in HemaSphere: June 2021 - Volume 5; 2021 Jun 9–17.
  • Kater AP, Kipps T, Eichhorst B, et al. Five-year analysis of Murano study demonstrates enduring undetectable minimal residual disease (uMRD) in a subset of relapsed/refractory chronic lymphocytic leukemia (R/R CLL) patients (pts) following fixed-duration venetoclax-rituximab (VenR) therapy (tx). American Society of Hematology Annual Meeting & Exposition (virtual), Blood (2020) 136 (Supplement 1): 19–21.
  • Kater AP, Seymour JF, Hillmen P, et al. Fixed duration of venetoclax-rituximab in relapsed/refractory chronic lymphocytic leukemia eradicates minimal residual disease and prolongs survival: post-treatment follow-up of the MURANO phase III study. J Clin Oncol. 2019;37(4):269–277.
  • Kater AP, Kersting S, van Norden Y, HOVON CLL Study Group, et al. Obinutuzumab pretreatment abrogates tumor lysis risk while maintaining undetectable MRD for venetoclax + obinutuzumab in CLL. Blood Adv. 2018;2(24):3566–3571.
  • Fischer K, Al-Sawaf O, Fink AM, et al. Venetoclax and obinutuzumab in chronic lymphocytic leukemia. Blood. 2017;129(19):2702–2705.
  • Ma S, Seymour JF, Brander DM, et al. Efficacy of venetoclax plus rituximab for relapsed CLL: five-year follow-up of continuous or limited-duration therapy. Blood. 2021;138(10):836–846.
  • Seymour JF, Ma S, Brander DM, et al. Venetoclax plus rituximab in relapsed or refractory chronic lymphocytic leukaemia: a phase 1b study. Lancet Oncol. 2017;18(2):230–240.
  • Munn Z, Peters MDJ, Stern C, et al. Systematic review or scoping review? Guidance for authors when choosing between a systematic or scoping review approach. BMC Med Res Methodol. 2018;18(1):143.
  • Ahn IE, Farooqui MZH, Tian X, et al. Depth and durability of response to ibrutinib in CLL: 5-year follow-up of a phase 2 study. Blood. 2018;131(21):2357–2366.
  • Wang XV, Hanson CA, Tschumper RC, et al. Measurable residual disease does not preclude prolonged progression-free survival in CLL treated with ibrutinib. Blood. 2021;138(26):2810–2827.
  • Fraser G, Cramer P, Demirkan F, et al. Updated results from the phase 3 HELIOS study of ibrutinib, bendamustine, and rituximab in relapsed chronic lymphocytic leukemia/small lymphocytic lymphoma. Leukemia. 2019;33(4):969–980.
  • Al-Sawaf O, Zhang C, Tandon M, et al. Fixed-duration venetoclax-obinutuzumab for previously untreated chronic lymphocytic leukemia: follow-up of efficacy and safety results from the multicenter, open-label, randomized phase 3 CLL14 trial. Abstract Book for the 25th Congress of the European Hematology Association Virtual Edition, 2020, in HemaSphere: June 2020 - Volume 4; 2020 Jun 11–21.
  • Al-Sawaf O, Zhang C, Tandon M, et al. Venetoclax plus obinutuzumab versus chlorambucil plus obinutuzumab for previously untreated chronic lymphocytic leukaemia (CLL14): follow-up results from a multicentre, open-label, randomised, phase 3 trial. Lancet Oncol. 2020;21(9):1188–1200.
  • Al-Sawaf O, Zhang C, Tandon M, CLL14 Study Investigators, et al. Fixed-duration venetoclax-obinutuzumab for previously untreated patients with chronic lymphocytic leukemia: Follow-up of efficacy and safety results from the multicenter, open-label, randomized, phase III CLL14 trial. J Clin Oncol. 2020;38(15_suppl):8027–8027.
  • Al-Sawaf O, Zhang C, Robrecht S, et al. Venetoclax-obinutuzumab for previously untreated chronic lymphocytic leukemia: 4-year follow-up analysis of the randomized CLL14 study. Hematol Oncol. 2021;39(S2):201–203.
  • Kater AP, Arslan O, Demikran F, et al. Efficacy of venetoclax in patients with relapsed/refractory chronic lymphocytic leukemia: primary endpoint analysis of the international phase 3b trial (VENICE I). Abstract Book for the 25th Congress of the European Hematology Association Virtual Edition, 2020, in HemaSphere: June 2020 - Volume 4; 2020 Jun 11–21.
  • Byrd JC, Wierda WG, Davids MS, et al. Durability of response to venetoclax (VEN) in patients with CLL relapsed/refractory to ibrutinib and/or idelalisib. J Clin Oncol. 2018;36(15_suppl):7512–7512.
  • Jones JA, Mato AR, Wierda WG, et al. Venetoclax for chronic lymphocytic leukaemia progressing after ibrutinib: an interim analysis of a multicentre, open-label, phase 2 trial. Lancet Oncol. 2018;19(1):65–75.
  • Stilgenbauer S, Eichhorst B, Schetelig J, et al. Venetoclax for patients with chronic lymphocytic leukemia with 17p deletion: results from the full population of a phase II pivotal trial. J Clin Oncol. 2018;36(19):1973–1980.
  • Kater AP, Wu JQ, Kipps T, et al. Venetoclax plus rituximab in relapsed chronic lymphocytic leukemia: 4-year results and evaluation of impact of genomic complexity and gene mutations from the MURANO phase III study. J Clin Oncol. 2020;38(34):4042–4054.
  • Munir T, Moreno C, Owen C, et al. First prospective data on minimal residual disease (MRD) outcomes after fixed-duration ibrutinib plus venetoclax (ibr + ven) versus chlorambucil plus obinutuzumab (clb + o) for first-line treatment of CLL in elderly or unfit patients: the glow study. Blood. 2021;138(Supplement 1):70–70.
  • Abrisqueta P, Gonzalez Barca E, Ferra CM, et al. Preliminary results of ibrutinib followed by ofatumumab consolidation in previously untreated patients with chronic lymphocytic leukemia (CLL): GELLC7 trials from the Spanish group of CLL (GELLC). Blood. 2019;134(Supplement_1):4296–4296.
  • Burger JA, Sivina M, Jain N, et al. Randomized trial of ibrutinib vs ibrutinib plus rituximab in patients with chronic lymphocytic leukemia. Blood. 2019;133(10):1011–1019.
  • Sharman JP, Farber CM, Mahadevan D, et al. Ublituximab (TG-1101), a novel glycoengineered anti-CD20 antibody, in combination with ibrutinib is safe and highly active in patients with relapsed and/or refractory chronic lymphocytic leukaemia: results of a phase 2 trial. Br J Haematol. 2017;176(3):412–420.
  • Lipsky AH, Hill BT, Winter AM, et al. High rates of undetectable minimal residual disease remissions with time-limited bendamustine, rituximab, and venetoclax (BR-VR) in untreated chronic lymphocytic leukemia (CLL). Blood. 2021;138(Supplement 1):1555–1555.
  • Wu JQ, Seymour JF, Eichhorst B, et al. Unfavorable genetics impact MRD response to venetoclax + rituximab retreatment in relapsed or refractory chronic lymphocytic leukemia (R/R CLL): phase 3 MURANO substudy. Hematol Oncol. 2021;39(S2):203–205.
  • Jain N, Ferrajoli A, Yilmaz M, et al. Venetoclax, obinutuzumab and atezolizumab (PD-L1 checkpoint inhibitor) for first-line treatment for patients with chronic lymphocytic leukemia (CLL). Blood. 2021;138(Supplement 1):2626–2626.
  • Staber PB, Jurczak W, Greil R, et al. Tafasitamab combined with idelalisib or venetoclax in patients with CLL previously treated with a BTK inhibitor. Leuk Lymphoma. 2021;62(14):3440–3451.
  • Hillmen P, Rawstron AC, Brock K, et al. Ibrutinib plus venetoclax in relapsed/refractory chronic lymphocytic leukemia: the CLARITY study. J Clin Oncol. 2019;37(30):2722–2729.
  • Wierda WG, Allan JN, Siddiqi T, et al. Ibrutinib plus venetoclax for first-line treatment of chronic lymphocytic leukemia: primary analysis results from the minimal residual disease cohort of the randomized phase II CAPTIVATE study. J Clin Oncol. 2021;39(34):3853–3865.
  • Niemann CU, Dubois J, Brieghel C, et al. Time-limited venetoclax and ibrutinib for patients with relapsed/refractory chronic lymphocytic leukemia (R/R CLL) who have undetectable MRD – primary analysis from the randomized phase II vision HO141 trial. Blood. 2021;138(Supplement 1):69–69.
  • von Tresckow J, Cramer P, Bahlo J, et al. CLL2-BIG: sequential treatment with bendamustine, ibrutinib and obinutuzumab (GA101) in chronic lymphocytic leukemia. Leukemia. 2019;33(5):1161–1172.
  • Michallet AS, Letestu R, Le Garff-Tavernier M, et al. A fixed-duration, measurable residual disease-guided approach in CLL: follow-up data from the phase 2 ICLL-07 FILO trial. Blood. 2021;137(8):1019–1023.
  • Michallet AS, Dilhuydy MS, Subtil F, et al. Obinutuzumab and ibrutinib induction therapy followed by a minimal residual disease-driven strategy in patients with chronic lymphocytic leukaemia (ICLL07 FILO): a single-arm, multicentre, phase 2 trial. Lancet Haematol. 2019;6(9):e470–e479.
  • Davids MS, Brander DM, Kim HT, Blood Cancer Research Partnership of the Leukemia & Lymphoma Society, et al. Ibrutinib plus fludarabine, cyclophosphamide, and rituximab as initial treatment for younger patients with chronic lymphocytic leukaemia: a single-arm, multicentre, phase 2 trial. Lancet Haematol. 2019;6(8):e419–e428.
  • Cramer P, Tresckow JV, Robrecht S, et al. Bendamustine, followed by ofatumumab and ibrutinib in chronic lymphocytic leukemia (CLL2-BIO): primary endpoint analysis of a multicenter, open-label phase-II trial. Haematologica. 2021;106(2):543–554.
  • Davids M, Fisher D, Tyekucheva S, et al. A phase Ib/II study of duvelisib in combination with FCR (DFCR) for frontline therapy of younger CLL patients. Abstract Book for the 23rd Congress of the European Hematology Association Stockholm, Sweden, June 14-17, 2018 in HemaSphere: June 2018 - Volume 2, page 351.
  • Coutre S, Choi M, Furman RR, et al. Venetoclax for patients with chronic lymphocytic leukemia who progressed during or after idelalisib therapy. Blood. 2018;131(15):1704–1711.
  • Sharman JP, Brander DM, Mato AR, et al. Ublituximab plus ibrutinib versus ibrutinib alone for patients with relapsed or refractory high-risk chronic lymphocytic leukaemia (GENUINE): a phase 3, multicentre, open-label, randomised trial. Lancet Haematol. 2021;8(4):e254–e266.
  • Sharman JP, Brander DM, Mato AR, et al. Effect of adding ublituximab to ibrutinib on PFS, ORR, and MRD negativity in previously treated high-risk chronic lymphocytic leukemia: final results of the GENUINE phase III study. J Clin Oncol. 2020;38(15_suppl):8022–8022.
  • Levin M-D, Dubois J, Nasserinejad K, et al. MRD-guided or fixed 12 cycles of venetoclax consolidation after venetoclax plus obinutuzumab treatment in first-line FCR unfit patients with CLL: primary endpoint analysis of the HOVON 139/GIVE trial. HemaSphere. 2021;5(S2):29.
  • Levin MD, Kersting S, Van Norden Y, et al. Obinutuzumab pre-induction abrogates high tumor lysis risk of venetoclax in treatment naive FCR-unfit patients with chronic lymphocytic leukemia: a planned interim analysis. Abstract Book for the 23rd Congress of the European Hematology Association Stockholm, Sweden, June 14-17, 2018 in HemaSphere: June 2018 - Volume 2: page 126.
  • Cramer P, von Tresckow J, Bahlo J, et al. Bendamustine followed by obinutuzumab and venetoclax in chronic lymphocytic leukaemia (CLL2-BAG): primary endpoint analysis of a multicentre, open-label, phase 2 trial. Lancet Oncol. 2018;19(9):1215–1228.
  • Wu J, Bolen C, Seymour JF, et al. Impact of major genomic alterations on outcome of relapsed/refractory chronic lymphocytic leukemia patients receiving venetoclax plus rituximab in the phase 3 MURANO study. Hematol Oncol. 2019;37(S2):106–108.
  • Kater A, Owen C, Moreno C, et al. Fixed-duration ibrutinib and venetoclax (I + V) versus chlorambucil plus obinutuzumab (clb + O) for first-line (1L) chronic lymphocytic leukemia (CLL): primary analysis of the phase 3 GLOW study. Abstract Book for the 26th Congress of the European Hematology Association Virtual Edition, 2021, in HemaSphere: June 2021 - Volume 5 Jun 9–17.
  • Ghia P, Allan JN, Siddiqi T, et al. Fixed-duration (FD) first-line treatment (tx) with ibrutinib (I) plus venetoclax (V) for chronic lymphocytic leukemia (CLL)/small lymphocytic lymphoma (SLL): primary analysis of the FD cohort of the phase 2 captivate study. J Clin Oncol. 2021;39(15_suppl):7501–7501.
  • Tam CSL, Allan JN, Siddiqi T, et al. CAPTIVATE primary analysis of first-line treatment with fixed-duration ibrutinib plus venetoclax for chronic lymphocytic leukemia (CLL)/small lymphocytic lymphoma (SLL. Hematol Oncol. 2021;39(S2):66–68.
  • Jain N, Keating M, Thompson P, et al. Ibrutinib plus venetoclax for first-line treatment of chronic lymphocytic leukemia: a nonrandomized phase 2 trial. JAMA Oncol. 2021;7(8):1213–1219.
  • Davids MS, Lampson BL, Tyekucheva S, et al. Updated safety and efficacy results from a phase 2 study of acalabrutinib, venetoclax and obinutuzumab (AVO) for frontline treatment of chronic lymphocytic leukemia (CLL). Blood. 2020;136(Supplement 1):20–21.
  • Davids MS, Lampson BL, Tyekucheva S, et al. Acalabrutinib, venetoclax, and obinutuzumab as frontline treatment for chronic lymphocytic leukaemia: a single-arm, open-label, phase 2 study. Lancet Oncol. 2021;22(10):1391–1402.
  • Baliakas P, Jeromin S, Iskas M, ERIC, the European Research Initiative on CLL, et al. Cytogenetic complexity in chronic lymphocytic leukemia: definitions, associations, and clinical impact. Blood. 2019;133(11):1205–1216.
  • Wu JQ, Seymour JF, Eichhorst B, et al. Impact of unfavorable genetics on minimal residual disease (MRD) response to venetoclax + rituximab retreatment in relapsed or refractory chronic lymphocytic leukemia (R/R CLL): phase 3 MURANO substudy. HemaSphere. 2021;5(S2):265–266.
  • Rogers KA, Huang Y, Ruppert AS, et al. Phase II study of combination obinutuzumab, ibrutinib, and venetoclax in treatment-naïve and relapsed or refractory chronic lymphocytic leukemia. J Clin Oncol. 2020;38(31):3626–3637.
  • Jain N, Thompson PA, Burger JA, et al. Ibrutinib, fludarabine, cyclophosphamide, and obinutuzumab (iFCG) for first-line treatment of IGHV-mutated CLL and without del(17p)/mutated TP53. Blood. 2019;134(Supplement_1):357–357.
  • Jain N, Thompson P, Burger J, et al. Ibrutinib, fludarabine, cyclophosphamide, and obinutuzumab (iFCG) regimen for chronic lymphocytic leukemia (CLL) with mutated IGHV and without TP53 aberrations. Leukemia. 2021;35(12):3421–3429.
  • Rawstron A, Webster N, Dalal S, et al. Kinetics of response in the peripheral blood predicts long-term responses to ibrutinib + venetoclax treatment for relapsed/refractory CLL in the Bloodwise TAP Clarity trial. Abstract Book for the 25th Congress of the European Hematology Association Virtual Edition, 2020, in HemaSphere: June 2020 - Volume 4; 2020 Jun 11–21.
  • Davids MS, Brander DM, Tyekucheva S, et al. Longer term follow-up of a multicenter, phase 2 study of ibrutinib plus fludarabine, cyclophosphamide, rituximab (iFCR) as initial therapy for younger patients with chronic lymphocytic leukemia. Blood. 2021;138(Supplement 1):640–640.
  • Roeker LE, Leslie LA, Soumerai JD, et al. A phase 2 study evaluating the addition of ublituximab and umbralisib (U2) to ibrutinib in patients with chronic lymphocytic leukemia (CLL): a minimal residual disease (MRD)-driven, time-limited approach. Blood. 2021;138(Supplement 1):395–395.
  • Al-Sawaf O, Zhang C, Lu T, et al. Venetoclax-obinutuzumab modulates clonal growth: results of a population-based minimal residual disease model from the randomized CLL14 study. HemaSphere. 2021;5(S2):282.
  • Jain N, Keating MJ, Thompson PA, et al. Combined ibrutinib and venetoclax for first-line treatment of patients with chronic lymphocytic leukemia (CLL): focus on long-term MRD results. Blood. 2020;136(Supplement 1):42–43.
  • Thompson PA, Wang Y, Keating MJ, et al. Venetoclax consolidation in patients with high-risk CLL who have been on ibrutinib more than a year achieves a high rate of undetectable minimal residual disease. Blood. 2021;138(Supplement 1):3723–3723.
  • Barr PM, Hill BT, Ma S, et al. A phase 1/2 study of umbralisib ublituximab and venetoclax in patients with relapsed or refractory chronic lymphocytic leukemia (CLL). Blood. 2019;134(Supplement_1):360–360.
  • Barr PM, Ma S, Zent CS, et al. 1/2 Study of umbralisib, ublituximab, and venetoclax (U2-Ven) in patients with relapsed or refractory chronic lymphocytic leukemia (CLL. American Society of Hematology Annual Meeting & Exposition (Virtual)Abstract Book for the 25th Congress of the European Hematology Association Virtual Edition, 2020, in HemaSphere: June 2020 - Volume 4; 2020 Dec 7–11
  • Scarfò L, Heltai S, Albi E, et al. Minimal residual disease-driven treatment intensification with sequential addition of ibrutinib (IBR) to venetoclax (VEN) in relapsed/refractory chronic lymphocytic leukemia (CLL): the IMPROVE study. HemaSphere. 2021;5(S2):283–284.
  • Al-Sawaf O, Zhang C, Robrecht S, et al. Clonal dynamics after venetoclax-obinutuzumab therapy: novel insights from the randomized, phase 3 CLL14 trial. Blood. 2020;136(Supplement 1):22–23.
  • Hallek M, Cheson BD, Catovsky D, et al. iwCLL guidelines for diagnosis, indications for treatment, response assessment, and supportive management of CLL. Blood. 2018;131(25):2745–2760.
  • Letestu R, Cartron G, Lepretre S, et al. Minimal residual disease (MRD) by 8-color flow cytometry (Flow-MRD) and IGH clonospecific quantitative PCR (ASO RQPCR) reached similar performances for evaluation of CLL treatment in a phase II clinical trial: cross validation of the methods. Blood. 2014;124(21):3307–3307.
  • Rawstron AC, Villamor N, Ritgen M, et al. International standardized approach for flow cytometric residual disease monitoring in chronic lymphocytic leukaemia. Leukemia. 2007;21(5):956–964.