Deep remission and prolonged disease-free survival can be achieved with first-line chemoimmunotherapy (CIT), such as combined fludarabine, cyclophosphamide, and rituximab, in the majority of patients with chronic lymphocytic leukemia (CLL). More modest results are reported with less intense regimens like obinutuzumab plus chlorambucil. Clinical assessment has limited sensitivity in detecting residual disease responsible for subsequent relapse, even including morphologic bone marrow (BM) evaluation. Multicolor flow cytometry and polymerase chain reaction (PCR)-based methods can detect minimal residual disease (MRD) to a sensitivity of ≥1:10 000 (10−4). Achieving BM MRD-negative complete remission (CR) is associated with superior progression-free survival (PFS) and overall survival; MRD status is the single best posttreatment predictor of long-term outcomes after CIT. Newer oral B-cell receptor signaling pathway inhibitors are highly effective at controlling disease, but best monotherapy responses are typically partial remission, and patients must remain on treatment to maintain disease control. Therapeutic progress is still needed for CLL. We propose that targeting MRD provides opportunity to realize this progress. Achieving BM MRD-negative CR is a prerequisite for long-term unmaintained disease-free survival and potential for cure. We review available methodologies for detecting MRD and correlations with posttreatment outcomes. We discuss the potential utility of MRD to direct individualized therapy. Finally, we discuss the importance of MRD-negative status as a surrogate marker for longer PFS in clinical studies to allow more rapid determination of clinical benefit.

Accurate quantitation of posttreatment residual disease burden in chronic lymphocytic leukemia (CLL) is prognostically relevant. Achievement of complete remission (CR) is associated with superior progression-free (PFS)1-6  and overall survival (OS) in first-line and relapsed/refractory CLL.1-3  Most patients who achieve CR have persistent, low-level disease, which is not detected with routine imaging and laboratory tests but can be demonstrated in bone marrow (BM) and/or blood with more sensitive flow cytometry (FLC) and molecular methods, so-called minimal residual disease (MRD). These residual cells ultimately are responsible for clinical relapse. The timing of relapse depends on the quantity of residual disease and kinetics of residual leukemia cell division. Sensitive methods to detect and quantify MRD were developed to more accurately predict subsequent clinical outcomes. Efforts to establish standards for MRD detection have implemented multiparameter FLC and polymerase chain reaction (PCR) assays with sensitivity to detect ≥1 CLL cell in 10 000 normal leukocytes (ie, a sensitivity of ≥10−4).7-9  Achieving MRD-negative remission is the single most important predictor of PFS3,10,11  and OS in patients treated with chemoimmunotherapy (CIT), independent of clinical remission status and pretreatment patient characteristics.

The ability to achieve deep remission depends on the therapeutic agents and strategy: chlorambucil monotherapy is palliative, with few CRs and limited effect on the natural history of CLL.12  However, durable, unmaintained CR is achieved in the majority of fit patients treated with fludarabine-based CIT regimens containing CD20 monoclonal antibody (mAb).2,6,13,14  CR can also be achieved in some elderly patients and those with comorbidities with chlorambucil combined with the CD20 mAbs obinutuzumab15  and ofatumumab.16 

The therapeutic paradigm with small molecule inhibitors of the B-cell receptor (BCR) signaling pathway is distinct. The BCR signaling pathway inhibitors ibrutinib17-19  and idelalisib20  achieve rapid and dramatic nodal reduction and improvement in hemoglobin and platelet count. Partial response is achieved by most patients treated with these oral agents when used for relapsed/refractory CLL, most often due to persistence of bone marrow (BM) disease; MRD-negative CR is rare. Long-term continuous therapy with these agents is required to achieve durable disease control.19  The CR rate for ibrutinib monotherapy is higher in the first-line setting,19,21  but MRD-negative CR remains rare.

To date, MRD testing has focused predominantly on its ability to predict clinical outcomes in clinical trials. We speculate that the future will see MRD-directed, individualized therapeutic approaches, for the following reasons: first, widespread availability of highly sensitive MRD testing; second, the association between MRD-negative remission and improved clinical outcomes; finally, the development of therapeutic strategies to achieve MRD-negative remissions in higher numbers of patients with reduced toxicity, such that a higher proportion of patients will be suitable for therapy designed to achieve deep remissions. Such approaches may allow attenuating treatment based on early response and the use of consolidation therapies in patients with persistent MRD positivity and/or high-risk disease biology.

In this review, we discuss available MRD testing methods, existing clinical data using these methods, and how MRD testing may be further developed and applied in the future using existing and novel therapeutic strategies.

In CLL, an ideal MRD assay should have the following characteristics: quantitative, with specific levels predictive of time-to-clinical events; standardized methodology and interpretation of laboratory data, to allow comparison between laboratories and clinical trials; broadly applicable, regardless of pretreatment biological characteristics; high throughput, with rapid turnaround time; and relatively noninvasive, technically simple, with objective and easy to interpret results.

CLL is a multicompartmental disease, nearly always involving BM, blood, lymph nodes, liver, and spleen (macroscopically or microscopically) prior to treatment. Following treatment, 1 or more of these disease sites may act as a “reservoir” for residual disease. As such, MRD testing in CLL is potentially more complex and challenging than for other leukemias, because current methodology focuses on sampling low-level disease from blood or BM, which is likely limiting.

Table 1 summarizes the methods used for detecting MRD in CLL.22  Standardization currently requires an acceptable MRD assay to have a sensitivity of ≥10−4 mononuclear cells (0.01% or 10−4), regardless of site of evaluation.

Flow cytometry

Basic 3-color FLC for CLL detects CD5/19 coexpression and κ/λ light chain restriction. It is inadequate because sensitivity approximates 1% (10−3), limited by the presence of normal CD5/19+ B cells and difficulty demonstrating monoclonality when B-cell count is low.23-26 

Rawstron et al standardized and optimized a 4-color FLC technique with sensitivity of ≥10−4 (0.01%), consisting of multicolor mAb panels to detect antigens differentially expressed on CLL vs normal B and T cells. It does not require patient-specific reagents and can be performed in most clinical FLC laboratories.7  It is quantitative and can be performed on blood and BM.27  This method has been widely adopted in clinical trials and can be used in routine practice. There were initial concerns that rituximab-induced loss of CD20 expression would impair performance, because dimmer CD20 expression on CLL cells relative to normal B cells is a specific differentiating feature. However, due to normal B-cell depletion by rituximab, this does not affect assay performance in practice.28  Interpretation is, however, somewhat operator dependent, and it is less sensitive than molecular methods; the main determinant of sensitivity is the number of viable leukocytes analyzed. Sensitivity of 10−4 requires acquisition of ≥500 000 events; sensitivity may, therefore, be lower in hypocellular specimens.29  Taking advantage of technological advances, FLC methodology is being further refined; 6-color,8  8-color,29,30  and 10-color methods were developed.31  The latter is a single tube assay and sensitivity approaches 10−5 if 1.8 × 106 total cells are analyzed. Finally, addition of the tumor-specific antigen CD160 may achieve sensitivity of 0.01% to 0.001% in a 6-color, single-tube assay.32 

PCR-based molecular methodologies

PCR using consensus primers that bind to conserved regions of the immunoglobulin heavy chain variable (IGHV) gene, adjacent to the complementarity determining region 3 (CDR3), has limited sensitivity in detecting MRD; although as few as 2% CLL cells can be detected in a background of normal B cells, amplification of background polyclonal IGHV limits sensitivity to ∼10−2.27  The ability to identify the malignant clone in the background of polyclonal B cells depends on the uniqueness of the CDR3 in the CLL clone, with resultant interpatient variability in assay sensitivity.9  Finally, some CLL clones with mutated IGHV will not amplify due to failure of binding of consensus primers.

PCR using patient-specific primers achieves sensitivity of ∼10−5.9  The IGHV of the malignant clone is sequenced, and allele-specific oligonucleotide (ASO) probes designed for the specific IGHV CDR3 region. The PCR reaction specifically expands the malignant IGHV gene. Using real-time quantitative PCR (RQ-PCR) methodology, the assay is quantitative.33-35  If combined with a consensus PCR step (nested ASO IGHV PCR), sensitivity can be increased (∼10−6), but quantitative ability is lost.36  The quantitative range and sensitivity need to be determined for each individual patient; thus, it is labor intensive, expensive, and time-consuming.9  Advantages compared with flow cytometry include higher sensitivity, requirement for less material and the stability of DNA at room temperature, which allows testing of older samples.9  Standardized procedures have been adopted to improve interlaboratory concordance and reproducibility.35 

A novel PCR-based method uses high-throughput sequencing (HTS).37  The patient’s clonotype is sequenced at baseline. In posttreatment samples, consensus (rather than patient-specific) primers universally amplify IGHV gene segments, followed by performance of HTS; the malignant clone is quantified algorithmically within the polyclonal background, which means the assay does not require validation for individual patients.38  This methodology is highly sensitive (∼10−6, quantitative to 10−5). Cells from blood or BM can be evaluated. The assay can also detect cell-free DNA in plasma. In diffuse large B-cell lymphoma, a predominantly nodal disease, HTS for IGHV genes to detect cell-free tumor-associated DNA was both sensitive and specific for the early detection of relapsing disease.39  Plasma evaluation for cell-free DNA was 2 log more sensitive at detecting residual disease than testing the cellular fraction of blood.40,41  Cell-free DNA in plasma is likely present due to apoptosis/necrosis of tumor cells in the tissues.42  Thus far, HTS MRD testing in CLL has only been performed on the mononuclear cell fraction of blood.37  Nonetheless, testing of the plasma component for cell-free DNA may hold promise for assessing the presence of low-level residual disease in the extravascular compartments and its sensitivity is an important future research question.

MRD correlated with PFS and OS in studies using diverse treatments and MRD analysis strategies.

MRD assessment following combination chemotherapy and CIT in first-line treatment

Table 2 summarizes studies in treatment-naïve patients treated with combination chemotherapy or CIT, where MRD analysis with a sensitivity of ≥10−4 was performed.43-45  In the German CLL Study Group (GCLLSG) CLL8 study, the level of MRD in blood and BM at final response assessment correlated with both PFS and OS, independent of pretreatment patient characteristics and treatment group. However, the likelihood of achieving MRD-negative remission was lower in patients with high-risk biological features and in those treated with fludarabine and cyclophosphamide (FC) vs fludarabine, cyclophosphamide, and rituximab (FCR).10  Nevertheless, achieving MRD-negative status was an independent predictor of PFS after treatment with combination chemotherapy3,30  and both low-intensity15  and moderate intensity CIT regimens.10,46-48  Level of MRD was significantly associated with OS in all series where it was reported.10,46,49 

MRD assessment postsalvage treatment and in treatment of high-risk populations

Table 3 summarizes studies reporting clinical outcomes by MRD status in relapsed/refractory or high-risk populations. MRD-negative remission was associated with superior PFS after FCR treatment,5  in patients with TP53 deletion treated with alemtuzumab plus high-dose methylprednisolone,50  and with salvage alemtuzumab monotherapy,51  independent of clinical response category.

MRD assessment post–allogenic stem cell transplant

Studies showed that MRD-negative status at 12 months post–allogenic stem cell transplant (allo-SCT) was associated with superior event-free survival (EFS; Table 4), using 4-color FLC (sensitivity 10−4),52  nested ASO IGHV PCR assay (sensitivity 10−6-10−7),53  and HTS-based consensus IGHV PCR (sensitivity 10−6).37  Logan et al compared the predictive ability of the HTS PCR assay vs 4-color FLC: EFS at 50 months after allo-SCT was 93.3% vs 37.5% for patients with undetectable vs detectable MRD assessed by PCR and 86% vs 20% when assessed by FLC. The higher sensitivity of the HTS PCR assay marginally improved the negative predictive value but reduced the positive predictive value of MRD testing for relapse. Significant fluctuation in MRD levels was seen up to the 12-month time point, at which point most patients had successfully been weaned off immunosuppression.37 

Given that allo-SCT has curative potential and intent, 1 major aim for MRD monitoring with this treatment approach might be to direct preemptive intervention, particularly with immunomodulation (withdrawal of immunosuppression ± donor lymphocyte infusion), when disease burden is low and the intervention potentially has maximal efficacy. To date, no studies specifically analyzed the efficacy of MRD-directed intervention. In the GCLLSG CLL3X study,52  patients who were MRD negative had lower relapse risk and superior EFS; MRD-directed preemptive donor lymphocyte infusion achieved MRD-negative CR in 3 of 6 patients. Patient number was small, and interventions were not protocol mandated; the role of MRD-directed, preemptive immunomodulatory therapy requires systematic investigation.

MRD-negative remission after CIT is highly correlated with PFS and OS. In the original phase 2 MD Anderson FCR cohort,2,14  a ligase-based PCR assay for clonal IGHV was performed at final response assessment.54  This assay achieved greater discrimination of outcomes than clinical assessment of remission status alone; in patients who achieved CR, median PFS was 13.7 years in MRD-negative patients vs 6.2 years in MRD-positive patients (P < .0001). Patients with mutated IGHV who were MRD negative had a PFS of almost 80% at 13 years.55  The potential to achieve highly prolonged, disease-free survival after first-line therapy, in a subgroup of patients, places additional importance on MRD as an end point, because achieving MRD-negative CR is a prerequisite for such an outcome.

Potential benefits of achieving MRD-negative status and long-term remission without maintenance therapy include avoidance of potential for cumulative toxicity from maintenance or sequential therapies and reduction in the financial burden on the patient and society through a delay in the use of expensive targeted therapies. Achieving MRD-negative status may reduce the likelihood of developing resistance mutations, although this remains to be proven. Finally, CLL cells induce immune dysfunction,56  which leads to increased susceptibility to infection, autoimmunity, and potentially other cancers57 ; an important future question is whether achieving deep remissions can reverse this immune dysfunction. However, the beneficial effects on immune function from achieving MRD-negative status must be balanced with the toxicity of the therapy required to achieve this. Fludarabine-based CIT, the therapy most able to achieve MRD-negative status, is associated with short and long-term immunosuppression, related to neutropenia and profound lymphopenia. In addition, long-term toxicity must be considered, such as the development of secondary myelodysplasia and acute myeloid leukemia. The only therapy shown to potentially enhance immune reconstitution is lenalidomide, which is associated with increases in immunoglobulin levels with monotherapy58  and improvements in T-cell function in vitro.59,60 

Recently, several potent novel therapies have shown potential to achieve MRD-negative remission with modest toxicity, bringing this goal potentially within reach of the majority of patients. The type II CD20 mAb obinutuzumab15  combined with chlorambucil achieved MRD-negative status in 19.5% of patients in BM and 37.5% in blood. Patients were elderly or had comorbidities and the regimen was well tolerated. Long-term follow-up of a phase 2 study of lenalidomide monotherapy as first-line treatment in older patients showed that MRD-negative CR was achieved in 8% of patients.47  The B-cell CLL/lymphoma 2 (BCL-2) inhibitor venetoclax can also achieve MRD-negative remission in relapsed/refractory patients, as monotherapy; 22% achieved CR/CR with incomplete marrow recovery (CRi); 5 of 9 CR/CRi patients tested were MRD negative.61  A phase 1 dose escalation study of venetoclax plus rituximab demonstrated MRD-negative BM in 24 (49%) of 49 patients.62  As experience is gained with ibrutinib and ibrutinib plus rituximab in the first-line setting, it will be important to note if extended treatment is associated with continued deepening of responses and achievement of MRD-negative state, analogous to that seen in chronic myeloid leukemia treated with imatinib,63  and whether this may allow study of treatment cessation in some patients. Combinations of novel therapies are likely to considerably increase the proportion of patients who achieve MRD-negative remission. For example, there is in vitro synergy between ibrutinib and venetoclax64 ; a study is planned at our institution with this combination and a study with a combination of ibrutinib, venetoclax and obinutuzumab is ongoing (NCT02427451). Whether achieving MRD-negative remisson with such combinations results in durable, unmaintained remission analogous to that seen after CIT will be an important future question, as will the impact of pretreatment disease characteristics on duration of such remissions.

What is the optimal site to test?

Few studies directly compared MRD analysis from blood and BM. Abrisqueta et al showed that 12 of 57 (21%) patients treated with fludarabine, cytarabine, mitoxantrone, and rituximab were MRD negative in blood but remained MRD positive in BM.46  Several studies also reported results from both blood and BM in nonpaired analyses. In the GCLLSG CLL8 study, 63% of patients were MRD negative in blood at final response and 44% in BM10 ; BM assessment was only performed to confirm clinical/radiologic CR, enriching the tested population for patients most likely to have achieved MRD-negative status. In patients treated with bendamustine and rituximab, 57.8% and 29.2% were MRD negative after treatment in blood and BM, respectively.46  Finally, after low-intensity CIT with obinutuzumab and chlorambucil, 20% were MRD-negative in BM and 38% in blood. Again, these were nonpaired specimens, with BM done to confirm CR. Thus, MRD is more likely to be present in BM than blood at completion of CIT.

The site of MRD sampling may significantly affect the predictive ability of the test. Fisher et al showed after treatment with bendamustine and rituximab, the median PFS was significantly shorter for a given level of MRD measured from blood compared with BM (PFS not reached [NR] vs 32.4 vs 11.7 months from blood compared with NR vs NR vs 29.7 months from BM for MRD levels of <10−4, ≥10−4 to <10−2 and ≥10−2, respectively). Thus, detection of MRD in blood was associated with short response duration; patients with MRD in blood after treatment may therefore represent an appropriate group in whom to study novel consolidation strategies. For treatment delivered with curative intent (eg, post–allo-SCT), MRD-negative CR is likely essential. Given higher rates of MRD after treatment in BM, assessment of BM MRD would achieve the highest negative predictive value for predicting subsequent relapse risk. In contrast, if the primary purpose of the assay is to identify poor-risk patients who may benefit from consolidation and/or maintenance therapy, then blood assessment may suffice.

While MRD testing can directly assess cells in blood and BM, residual disease in lymph nodes, liver, and spleen cannot be directly or easily assessed by these methods. Computed tomography and positron emission tomography have limited sensitivity and specificity for detecting small volume disease. HTS-based testing of plasma, as discussed earlier, may be evaluated in the future to detect MRD in these sites.

Impact of pretreatment biological characteristics on interpretation of MRD results

Achieving MRD-negative remission is associated with superior PFS and OS, independent of pretreatment prognostic features.10  However, the likelihood of achieving MRD-negative remission is lower for patients with high-risk prognostic features and those receiving less effective therapy. Additionally, in multivariable analysis, pretreatment high-risk characteristics, such as unmutated IGHV, ZAP70 positivity, and del(11q), were associated with more rapid reappearance of MRD and shorter PFS.3,65 

Disparities between clinical staging and MRD analysis

In the GCLLSG CLL8 study, 39% and 28% of patients who remained in partial remission (PR) by clinical criteria were MRD negative in blood and BM, respectively, at final response assessment.10  Updated analysis showed no difference in PFS between patients in MRD-negative CR vs MRD-negative PR; patients in MRD-negative PR had superior PFS to those in MRD-positive CR.66  Thus, achieving posttreatment MRD-negative remission with CIT may be more important than achieving clinical CR. In first-line treatment with obinutuzumab plus chlorambucil, 38% of patients in CR were MRD negative in blood compared with 31% of patients in PR.15  Similar results were seen in patients treated with venetoclax plus rituximab, where 50% of patients in PR and 70% of patients in CR were MRD negative.62  The existence of MRD-negative partial remission appears contradictory. However, there are 2 potential explanations for this phenomenon: first, residual lymph nodes of >1.5 cm by computed tomography, in the setting of MRD-negative remission, may not contain viable tumor (eg, with residual enlargement instead representing fibrosis). Second, lymph node–resident CLL cells may be more resistant to certain therapies, allowing persistence of viable tumor in lymph nodes, despite clearance of CLL from the blood and marrow. Biopsy of enlarged lymph nodes or novel techniques to detect MRD are needed to determine the clinical relevance of residual nodal enlargement in such patients who are MRD negative by FLC.

One proposed goal of MRD assessment is to develop risk-adapted treatment strategies, which requires prospective testing in clinical trial. Patients with MRD at final response assessment could be candidates for treatment intensification, consolidation, or maintenance strategies. Furthermore, patients who achieve early MRD-negative status may be candidates for treatment de-escalation to limit treatment-related toxicity.

Consolidation treatment

Consolidation therapy treats low-level residual disease, expected to otherwise produce clinical relapse, with non–cross-resistant agents, to achieve a greater depth of response. Treatment at a time of lower disease burden could reduce likelihood of developing drug resistance.

Studies evaluated the CD52 mAb alemtuzumab in this setting. Alemtuzumab improved PFS after fludarabine or FC chemotherapy, but was associated with significant infectious toxicity.67,68  Alemtuzumab consolidation after fludarabine and rituximab improved response: 61% of patients in PR achieved CR (50% MRD negative) and 43% of patients with MRD-positive CR became MRD negative. However, infection-related mortality was unacceptably high; consequently, no PFS or OS benefit was seen. Thus, consolidation with alemtuzumab after CIT is not recommended.69  Lenalidomide consolidation after CIT with pentostatin, fludarabine, and cyclophosphamide70  improved responses in 24% of patients with measurable disease; 4 became MRD negative, and time to next treatment appeared prolonged relative to historic data. However, many patients required dose reduction, particularly due to hematologic toxicity.71  The Cancer and Leukemia Group B 10404 study will further clarify the utility of lenalidomide consolidation after fludarabine-based CIT.

Toxicity of any consolidation therapy must be balanced against the benefit of improving depth of response, as the experience with alemtuzumab showed. Availability of potent novel therapies with more favorable toxicity profiles may lead to reexamination of the feasibility of MRD-directed consolidation after CIT. Venetoclax61,62  and obinutuzumab15  can induce MRD-negative CR and would likely be well tolerated as consolidation therapy. In addition, other novel therapies, such as immune checkpoint inhibitor mAbs and cellular therapy, are likely to be tested in this setting in the future.

MRD-guided therapy based on interim-analysis of MRD levels

No published study has prospectively used interim MRD levels to guide therapeutic decisions. In the GCLLSG CLL8 study, MRD analysis was performed in blood after 3 cycles of FC/FCR; all patients subsequently received 6 cycles, regardless of interim MRD results; and those achieving MRD-negative status had similar PFS, regardless of whether 3 or 6 cycles were required to achieve MRD-negative remission.10  Strati et al reported MRD results in treatment-naïve patients treated with FCR: MRD analysis was performed after 3 cycles and at completion of therapy; 17% and 43% of patients achieved MRD-negative status in BM after 3 and 6 cycles, respectively. Those who achieved MRD-negative status after 3 cycles and stopped treatment, most commonly due to therapy-related toxicity, had similar PFS as those who were MRD negative after 3 cycles but received the full 6 planned cycles and also to those who were MRD positive after 3 cycles but achieved MRD-negative status after 6 cycles.48  Taken together, these data suggest that early treatment cessation in patients who achieve MRD-negative status after 3 cycles of FCR may be feasible without compromising long-term disease control, but this requires prospective study.

Given the prolonged PFS after CIT, very long follow-up is required to determine whether PFS after receiving novel agents or regimens is superior to existing CIT regimens; this may significantly delay new drug development. Given the strong correlation between achieving MRD-negative status and survival outcomes, MRD status is receiving consideration from regulatory agencies as a potentially meaningful end point for clinical trials. A major caveat to this is that MRD assessment cannot generally be used to assess the efficacy of novel regimens such as ibrutinib as monotherapy or in combination with rituximab, given that these regimens rarely achieve MRD-negative status and are given continuously until disease progression.17-19,72 

Existing treatment paradigms in CLL are undergoing significant change. Deep remissions are achieved in a large number of fit patients receiving first-line therapy, and those patients with mutated IGHV potentially are treated with curative intent. Many other patients may achieve prolonged treatment-free remissions. Development of rational combinations and sequencing with novel therapies will likely further increase the number of patients achieving such remissions. The availability of sensitive and specific methods to quantify residual disease may allow individualized therapy in the future. Patients who achieve early MRD-negative status may have treatment de-escalated to limit toxicity, whereas those initially failing to achieve MRD-negative remission could receive consolidation or maintenance therapy with non–cross-resistant agents in the setting of low disease bulk, aiming to achieve an MRD-negative state, at which point treatment could be stopped with careful monitoring. For this to be a reality, highly sensitive and specific MRD detection methodology must be available. Whereas multicolor FLC is the current standard, more sensitive methods such as IGHV-HTS may ultimately prove to have superior predictive power; however, this will need to be determined prospectively.

P.A.T. received funding from the CLL Global Research Foundation and the Haematology Society of Australia and New Zealand.

Contribution: P.A.T. and W.G.W. reviewed and discussed important data and cowrote the paper.

Conflict-of-interest disclosure: W.G.W. is an advisor/consultant for Sanofi, Genentech/Roche, Pharmacyclics, Celgene, Gilead, GSK/Novartis, Genzyme, Merck, Abbvie, and Emergent and has contracted research from GSK/Novartis, Abbvie, Genentech, Karyopharm, Pharmacyclics, Ascerta, Gilead, Jansen, Emergent, Juno Therapeutics, and KITE Pharma. P.A.T. declares no competing financial interests.

Correspondence: William G. Wierda, Department of Leukemia, 1515 Holcombe Blvd, Unit 428, Houston, TX 77030; e-mail: wwierda@mdanderson.org.

1
Hallek
 
M
Fischer
 
K
Fingerle-Rowson
 
G
, et al. 
International Group of Investigators; German Chronic Lymphocytic Leukaemia Study Group
Addition of rituximab to fludarabine and cyclophosphamide in patients with chronic lymphocytic leukaemia: a randomised, open-label, phase 3 trial.
Lancet
2010
, vol. 
376
 
9747
(pg. 
1164
-
1174
)
2
Tam
 
CS
O’Brien
 
S
Wierda
 
W
, et al. 
Long-term results of the fludarabine, cyclophosphamide, and rituximab regimen as initial therapy of chronic lymphocytic leukemia.
Blood
2008
, vol. 
112
 
4
(pg. 
975
-
980
)
3
Bosch
 
F
Ferrer
 
A
Villamor
 
N
, et al. 
Fludarabine, cyclophosphamide, and mitoxantrone as initial therapy of chronic lymphocytic leukemia: high response rate and disease eradication.
Clin Cancer Res
2008
, vol. 
14
 
1
(pg. 
155
-
161
)
4
Hillmen
 
P
Skotnicki
 
AB
Robak
 
T
, et al. 
Alemtuzumab compared with chlorambucil as first-line therapy for chronic lymphocytic leukemia.
J Clin Oncol
2007
, vol. 
25
 
35
(pg. 
5616
-
5623
)
5
Wierda
 
W
O’Brien
 
S
Wen
 
S
, et al. 
Chemoimmunotherapy with fludarabine, cyclophosphamide, and rituximab for relapsed and refractory chronic lymphocytic leukemia.
J Clin Oncol
2005
, vol. 
23
 
18
(pg. 
4070
-
4078
)
6
Kay
 
NE
Geyer
 
SM
Call
 
TG
, et al. 
Combination chemoimmunotherapy with pentostatin, cyclophosphamide, and rituximab shows significant clinical activity with low accompanying toxicity in previously untreated B chronic lymphocytic leukemia.
Blood
2007
, vol. 
109
 
2
(pg. 
405
-
411
)
7
Rawstron
 
AC
Villamor
 
N
Ritgen
 
M
, et al. 
International standardized approach for flow cytometric residual disease monitoring in chronic lymphocytic leukaemia.
Leukemia
2007
, vol. 
21
 
5
(pg. 
956
-
964
)
8
Rawstron
 
AC
Böttcher
 
S
Letestu
 
R
, et al. 
European Research Initiative in CLL
Improving efficiency and sensitivity: European Research Initiative in CLL (ERIC) update on the international harmonised approach for flow cytometric residual disease monitoring in CLL.
Leukemia
2013
, vol. 
27
 
1
(pg. 
142
-
149
)
9
Böttcher
 
S
Hallek
 
M
Ritgen
 
M
Kneba
 
M
The role of minimal residual disease measurements in the therapy for CLL: is it ready for prime time?
Hematol Oncol Clin North Am
2013
, vol. 
27
 
2
(pg. 
267
-
288
)
10
Böttcher
 
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
, vol. 
30
 
9
(pg. 
980
-
988
)
11
Strati
 
P
Keating
 
MJ
O’Brien
 
SM
, et al. 
Outcomes of first-line treatment for chronic lymphocytic leukemia with 17p deletion.
Haematologica
2014
, vol. 
99
 
8
(pg. 
1350
-
1355
)
12
Rai
 
KR
Peterson
 
BL
Appelbaum
 
FR
, et al. 
Fludarabine compared with chlorambucil as primary therapy for chronic lymphocytic leukemia.
N Engl J Med
2000
, vol. 
343
 
24
(pg. 
1750
-
1757
)
13
Byrd
 
JC
Rai
 
K
Peterson
 
BL
, et al. 
Addition of rituximab to fludarabine may prolong progression-free survival and overall survival in patients with previously untreated chronic lymphocytic leukemia: an updated retrospective comparative analysis of CALGB 9712 and CALGB 9011.
Blood
2005
, vol. 
105
 
1
(pg. 
49
-
53
)
14
Keating
 
MJ
O’Brien
 
S
Albitar
 
M
, et al. 
Early results of a chemoimmunotherapy regimen of fludarabine, cyclophosphamide, and rituximab as initial therapy for chronic lymphocytic leukemia.
J Clin Oncol
2005
, vol. 
23
 
18
(pg. 
4079
-
4088
)
15
Goede
 
V
Fischer
 
K
Busch
 
R
, et al. 
Obinutuzumab plus chlorambucil in patients with CLL and coexisting conditions.
N Engl J Med
2014
, vol. 
370
 
12
(pg. 
1101
-
1110
)
16
Hillmen
 
P
Robak
 
T
Janssens
 
A
, et al. 
COMPLEMENT 1 Study Investigators
Chlorambucil plus ofatumumab versus chlorambucil alone in previously untreated patients with chronic lymphocytic leukaemia (COMPLEMENT 1): a randomised, multicentre, open-label phase 3 trial.
Lancet
2015
, vol. 
385
 
9980
(pg. 
1873
-
1883
)
17
Byrd
 
JC
Brown
 
JR
O’Brien
 
S
, et al. 
RESONATE Investigators
Ibrutinib versus ofatumumab in previously treated chronic lymphoid leukemia.
N Engl J Med
2014
, vol. 
371
 
3
(pg. 
213
-
223
)
18
Byrd
 
JC
Furman
 
RR
Coutre
 
SE
, et al. 
Targeting BTK with ibrutinib in relapsed chronic lymphocytic leukemia.
N Engl J Med
2013
, vol. 
369
 
1
(pg. 
32
-
42
)
19
Byrd
 
JC
Furman
 
RR
Coutre
 
SE
, et al. 
Three-year follow-up of treatment-naïve and previously treated patients with CLL and SLL receiving single-agent ibrutinib.
Blood
2015
, vol. 
125
 
16
(pg. 
2497
-
2506
)
20
Furman
 
RR
Sharman
 
JP
Coutre
 
SE
, et al. 
Idelalisib and rituximab in relapsed chronic lymphocytic leukemia.
N Engl J Med
2014
, vol. 
370
 
11
(pg. 
997
-
1007
)
21
O’Brien
 
S
Furman
 
RR
Coutre
 
SE
, et al. 
Ibrutinib as initial therapy for elderly patients with chronic lymphocytic leukaemia or small lymphocytic lymphoma: an open-label, multicentre, phase 1b/2 trial.
Lancet Oncol
2014
, vol. 
15
 
1
(pg. 
48
-
58
)
22
Robak
 
T
Blonski
 
JZ
Gora-Tybor
 
J
, et al. 
Polish Leukemia Group (PALG CLL2)
Cladribine alone and in combination with cyclophosphamide or cyclophosphamide plus mitoxantrone in the treatment of progressive chronic lymphocytic leukemia: report of a prospective, multicenter, randomized trial of the Polish Adult Leukemia Group (PALG CLL2).
Blood
2006
, vol. 
108
 
2
(pg. 
473
-
479
)
23
Robertson
 
LE
Huh
 
YO
Butler
 
JJ
, et al. 
Response assessment in chronic lymphocytic leukemia after fludarabine plus prednisone: clinical, pathologic, immunophenotypic, and molecular analysis.
Blood
1992
, vol. 
80
 
1
(pg. 
29
-
36
)
24
Vuillier
 
F
Claisse
 
JF
Vandenvelde
 
C
, et al. 
Evaluation of residual disease in B-cell chronic lymphocytic leukemia patients in clinical and bone-marrow remission using CD5-CD19 markers and PCR study of gene rearrangements.
Leuk Lymphoma
1992
, vol. 
7
 
3
(pg. 
195
-
204
)
25
Lenormand
 
B
Bizet
 
M
Fruchart
 
C
, et al. 
Residual disease in B-cell chronic lymphocytic leukemia patients and prognostic value.
Leukemia
1994
, vol. 
8
 
6
(pg. 
1019
-
1026
)
26
Cabezudo
 
E
Matutes
 
E
Ramrattan
 
M
Morilla
 
R
Catovsky
 
D
Analysis of residual disease in chronic lymphocytic leukemia by flow cytometry.
Leukemia
1997
, vol. 
11
 
11
(pg. 
1909
-
1914
)
27
Rawstron
 
AC
Kennedy
 
B
Evans
 
PA
, et al. 
Quantitation of minimal disease levels in chronic lymphocytic leukemia using a sensitive flow cytometric assay improves the prediction of outcome and can be used to optimize therapy.
Blood
2001
, vol. 
98
 
1
(pg. 
29
-
35
)
28
Böttcher
 
S
Stilgenbauer
 
S
Busch
 
R
, et al. 
Standardized MRD flow and ASO IGH RQ-PCR for MRD quantification in CLL patients after rituximab-containing immunochemotherapy: a comparative analysis.
Leukemia
2009
, vol. 
23
 
11
(pg. 
2007
-
2017
)
29
Stehlíková
 
O
Chovancová
 
J
Tichý
 
B
, et al. 
Detecting minimal residual disease in patients with chronic lymphocytic leukemia using 8-color flow cytometry protocol in routine hematological practice.
Int J Lab Hematol
2014
, vol. 
36
 
2
(pg. 
165
-
171
)
30
Raponi
 
S
Della Starza
 
I
De Propris
 
MS
, et al. 
Minimal residual disease monitoring in chronic lymphocytic leukaemia patients. A comparative analysis of flow cytometry and ASO IgH RQ-PCR.
Br J Haematol
2014
, vol. 
166
 
3
(pg. 
360
-
368
)
31
Sartor
 
MM
Gottlieb
 
DJ
A single tube 10-color flow cytometry assay optimizes detection of minimal residual disease in chronic lymphocytic leukemia.
Cytometry B Clin Cytom
2013
, vol. 
84
 
2
(pg. 
96
-
103
)
32
Farren
 
TW
Giustiniani
 
J
Fanous
 
M
, et al. 
Minimal residual disease detection with tumor-specific CD160 correlates with event-free survival in chronic lymphocytic leukemia.
Blood Cancer J
2015
, vol. 
5
 pg. 
e273
 
33
Pfitzner
 
T
Engert
 
A
Wittor
 
H
, et al. 
A real-time PCR assay for the quantification of residual malignant cells in B cell chronic lymphatic leukemia.
Leukemia
2000
, vol. 
14
 
4
(pg. 
754
-
766
)
34
van der Velden
 
VH
Hochhaus
 
A
Cazzaniga
 
G
Szczepanski
 
T
Gabert
 
J
van Dongen
 
JJ
Detection of minimal residual disease in hematologic malignancies by real-time quantitative PCR: principles, approaches, and laboratory aspects.
Leukemia
2003
, vol. 
17
 
6
(pg. 
1013
-
1034
)
35
van der Velden
 
VH
Cazzaniga
 
G
Schrauder
 
A
, et al. 
European Study Group on MRD detection in ALL (ESG-MRD-ALL)
Analysis of minimal residual disease by Ig/TCR gene rearrangements: guidelines for interpretation of real-time quantitative PCR data.
Leukemia
2007
, vol. 
21
 
4
(pg. 
604
-
611
)
36
Voena
 
C
Ladetto
 
M
Astolfi
 
M
, et al. 
A novel nested-PCR strategy for the detection of rearranged immunoglobulin heavy-chain genes in B cell tumors.
Leukemia
1997
, vol. 
11
 
10
(pg. 
1793
-
1798
)
37
Logan
 
AC
Zhang
 
B
Narasimhan
 
B
, et al. 
Minimal residual disease quantification using consensus primers and high-throughput IGH sequencing predicts post-transplant relapse in chronic lymphocytic leukemia.
Leukemia
2013
, vol. 
27
 
8
(pg. 
1659
-
1665
)
38
Faham
 
M
Zheng
 
J
Moorhead
 
M
, et al. 
Deep-sequencing approach for minimal residual disease detection in acute lymphoblastic leukemia.
Blood
2012
, vol. 
120
 
26
(pg. 
5173
-
5180
)
39
Kurtz
 
DM
Green
 
MR
Bratman
 
SV
, et al. 
Non-invasive monitoring of diffuse large B-cell lymphoma by immunoglobulin high-throughput sequencing.
Blood
2015
, vol. 
125
 
24
(pg. 
3679
-
3687
)
40
Bain
 
BJ
Catovsky
 
D
The leukaemic phase of non-Hodgkin’s lymphoma.
J Clin Pathol
1995
, vol. 
48
 
3
(pg. 
189
-
193
)
41
Muringampurath-John
 
D
Jaye
 
DL
Flowers
 
CR
, et al. 
Characteristics and outcomes of diffuse large B-cell lymphoma presenting in leukaemic phase.
Br J Haematol
2012
, vol. 
158
 
5
(pg. 
608
-
614
)
42
Schwarzenbach
 
H
Hoon
 
DS
Pantel
 
K
Cell-free nucleic acids as biomarkers in cancer patients.
Nat Rev Cancer
2011
, vol. 
11
 
6
(pg. 
426
-
437
)
43
Lamanna
 
N
Jurcic
 
JG
Noy
 
A
, et al. 
Sequential therapy with fludarabine, high-dose cyclophosphamide, and rituximab in previously untreated patients with chronic lymphocytic leukemia produces high-quality responses: molecular remissions predict for durable complete responses.
J Clin Oncol
2009
, vol. 
27
 
4
(pg. 
491
-
497
)
44
Maloum
 
K
Settegrana
 
C
Chapiro
 
E
, et al. 
IGHV gene mutational status and LPL/ADAM29 gene expression as clinical outcome predictors in CLL patients in remission following treatment with oral fludarabine plus cyclophosphamide.
Ann Hematol
2009
, vol. 
88
 
12
(pg. 
1215
-
1221
)
45
Kwok
 
M
Rawstron
 
A
Varghese
 
A
, et al. 
Independent prognostic significance of minimal residual disease status in chronic lymphocytic leukaemia.
Lancet
2014
, vol. 
383
 
special issue
pg. 
S66
 
46
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
, vol. 
30
 
26
(pg. 
3209
-
3216
)
47
Abrisqueta
 
P
Villamor
 
N
Terol
 
MJ
, et al. 
Rituximab maintenance after first-line therapy with rituximab, fludarabine, cyclophosphamide, and mitoxantrone (R-FCM) for chronic lymphocytic leukemia.
Blood
2013
, vol. 
122
 
24
(pg. 
3951
-
3959
)
48
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
, vol. 
123
 
24
(pg. 
3727
-
3732
)
49
Moreno
 
C
Villamor
 
N
Colomer
 
D
, et al. 
Clinical significance of minimal residual disease, as assessed by different techniques, after stem cell transplantation for chronic lymphocytic leukemia.
Blood
2006
, vol. 
107
 
11
(pg. 
4563
-
4569
)
50
Pettitt
 
AR
Jackson
 
R
Carruthers
 
S
, et al. 
Alemtuzumab in combination with methylprednisolone is a highly effective induction regimen for patients with chronic lymphocytic leukemia and deletion of TP53: final results of the national cancer research institute CLL206 trial.
J Clin Oncol
2012
, vol. 
30
 
14
(pg. 
1647
-
1655
)
51
Moreton
 
P
Kennedy
 
B
Lucas
 
G
, et al. 
Eradication of minimal residual disease in B-cell chronic lymphocytic leukemia after alemtuzumab therapy is associated with prolonged survival.
J Clin Oncol
2005
, vol. 
23
 
13
(pg. 
2971
-
2979
)
52
Dreger
 
P
Döhner
 
H
Ritgen
 
M
, et al. 
German CLL Study Group
Allogeneic stem cell transplantation provides durable disease control in poor-risk chronic lymphocytic leukemia: long-term clinical and MRD results of the German CLL Study Group CLL3X trial.
Blood
2010
, vol. 
116
 
14
(pg. 
2438
-
2447
)
53
Farina
 
L
Carniti
 
C
Dodero
 
A
, et al. 
Qualitative and quantitative polymerase chain reaction monitoring of minimal residual disease in relapsed chronic lymphocytic leukemia: early assessment can predict long-term outcome after reduced intensity allogeneic transplantation.
Haematologica
2009
, vol. 
94
 
5
(pg. 
654
-
662
)
54
Jilani
 
I
Keating
 
M
Day
 
A
, et al. 
Simplified sensitive method for the detection of B-cell clonality in lymphoid malignancies.
Clin Lab Haematol
2006
, vol. 
28
 
5
(pg. 
325
-
331
)
55
Thompson
 
PA
Tam
 
CS
O'Brien
 
SM
, et al. 
 
Fludarabine, cyclophosphamide and rituximab achieves long-term disease-free survival in IGHV-mutated chronic lymphocytic leukemia [published online ahead of print October 22, 2015]. Blood. doi:10.1182/blood-2015-09-667675
56
Riches
 
JC
Ramsay
 
AG
Gribben
 
JG
Immune dysfunction in chronic lymphocytic leukemia: the role for immunotherapy.
Curr Pharm Des
2012
, vol. 
18
 
23
(pg. 
3389
-
3398
)
57
Falchi
 
L
Keating
 
M
Lerner
 
S
, et al. 
Other cancers in long-term survivors of chronic lymphocytic leukemia: incidence and prognostic relevance.
Blood
2014
, vol. 
124
 
21
pg. 
3323
 
58
Badoux
 
XC
Keating
 
MJ
Wen
 
S
, et al. 
Lenalidomide as initial therapy of elderly patients with chronic lymphocytic leukemia.
Blood
2011
, vol. 
118
 
13
(pg. 
3489
-
3498
)
59
Ramsay
 
AG
Clear
 
AJ
Fatah
 
R
Gribben
 
JG
Multiple inhibitory ligands induce impaired T-cell immunologic synapse function in chronic lymphocytic leukemia that can be blocked with lenalidomide: establishing a reversible immune evasion mechanism in human cancer.
Blood
2012
, vol. 
120
 
7
(pg. 
1412
-
1421
)
60
Ramsay
 
AG
Gribben
 
JG
Immune dysfunction in chronic lymphocytic leukemia T cells and lenalidomide as an immunomodulatory drug.
Haematologica
2009
, vol. 
94
 
9
(pg. 
1198
-
1202
)
61
Seymour
 
JF
Davids
 
MS
Pagel
 
JM
, et al. 
ABT-199 (GDC-0199) in relapsed/refractory (R/R) chronic lymphocytic leukemia (CLL) and small lymphocytic lymphoma (SLL): high complete- response rate and durable disease control.
J Clin Oncol
2014
, vol. 
32
 
15 suppl
pg. 
7015
 
62
Roberts
 
AW
Ma
 
S
Brander
 
DM
, et al. 
 
Venetoclax (ABT-199/GDC-0199) combined with rituximab induces deep responses in patients with relapsed/refractory chronic lymphocytic leukemia [abstract]. Haematologica. 2015;100(Suppl 1):154. Abstract S431
63
Hughes
 
TP
Hochhaus
 
A
Branford
 
S
, et al. 
IRIS investigators
Long-term prognostic significance of early molecular response to imatinib in newly diagnosed chronic myeloid leukemia: an analysis from the International Randomized Study of Interferon and STI571 (IRIS).
Blood
2010
, vol. 
116
 
19
(pg. 
3758
-
3765
)
64
Cervantes-Gomez
 
F
Lamothe
 
B
Woyach
 
JA
, et al. 
Pharmacological and protein profiling suggest ABT-199 as optimal partner with ibrutinib in chronic lymphocytic leukemia.
Clin Cancer Res
2015
, vol. 
21
 
16
(pg. 
3705
-
3715
)
65
Boettcher
 
S
Ritgen
 
M
Fischer
 
K
, et al. 
Minimal residual disease (MRD) re-growth kinetics are an independent predictor for progression free survival (PFS) in chronic lymphocytic leukemia (CLL) and are related to biologically defined CLL-subgroups: results from the CLL8 trial of the German CLL Study Group (GCLLSG).
Blood
2011
, vol. 
118
 
21
pg. 
1777
 
66
Kovacs
 
G
Boettcher
 
S
Bahlo
 
J
, et al. 
Value of minimal residual disease (MRD) negative status at response evaluation in chronic lymphocytic leukemia (CLL): combined analysis of two phase III studies of the German CLL Study Group (GCLLSG).
Blood
2014
, vol. 
124
 
21
pg. 
23
 
67
Wendtner
 
CM
Ritgen
 
M
Schweighofer
 
CD
, et al. 
German CLL Study Group (GCLLSG)
Consolidation with alemtuzumab in patients with chronic lymphocytic leukemia (CLL) in first remission--experience on safety and efficacy within a randomized multicenter phase III trial of the German CLL Study Group (GCLLSG).
Leukemia
2004
, vol. 
18
 
6
(pg. 
1093
-
1101
)
68
Schweighofer
 
CD
Ritgen
 
M
Eichhorst
 
BF
, et al. 
Consolidation with alemtuzumab improves progression-free survival in patients with chronic lymphocytic leukaemia (CLL) in first remission: long-term follow-up of a randomized phase III trial of the German CLL Study Group (GCLLSG).
Br J Haematol
2009
, vol. 
144
 
1
(pg. 
95
-
98
)
69
Lin
 
KI
Tam
 
CS
Keating
 
MJ
, et al. 
Relevance of the immunoglobulin VH somatic mutation status in patients with chronic lymphocytic leukemia treated with fludarabine, cyclophosphamide, and rituximab (FCR) or related chemoimmunotherapy regimens.
Blood
2009
, vol. 
113
 
14
(pg. 
3168
-
3171
)
70
Shanafelt
 
TD
Ramsay
 
AG
Zent
 
CS
, et al. 
Long-term repair of T-cell synapse activity in a phase II trial of chemoimmunotherapy followed by lenalidomide consolidation in previously untreated chronic lymphocytic leukemia (CLL).
Blood
2013
, vol. 
121
 
20
(pg. 
4137
-
4141
)
71
Shanafelt
 
T
Treatment of older patients with chronic lymphocytic leukemia: key questions and current answers.
Hematol Am Soc Hematol Educ Progr
2013
, vol. 
2013
 (pg. 
158
-
167
)
72
Burger
 
JA
Keating
 
MJ
Wierda
 
WG
, et al. 
Safety and activity of ibrutinib plus rituximab for patients with high-risk chronic lymphocytic leukaemia: a single-arm, phase 2 study.
Lancet Oncol
2014
, vol. 
15
 
10
(pg. 
1090
-
1099
)
Sign in via your Institution