Background: Small molecules targeting Bruton’s tyrosine kinase (BTK) and B-cell lymphoma-2 have become the standard of care for the treatment of chronic lymphocytic leukemia (CLL), replacing chemoimmunotherapy (CIT) in most clinical settings. Ongoing trials explore targeted combinations and minimal residual disease-driven treatment cessation. These dramatic shifts in the current and upcoming treatment landscape of CLL raise the need to reevaluate existing prognostic markers and develop novel ones. Summary: This review examines prognostic markers in CLL patients treated with standard and investigational targeted therapies. Specifically, initial treatment of TP53 aberrant patients with a BTK inhibitor can achieve 70% progression-free survival (PFS) at 5 years, outperforming the 15% 5-year PFS with a CIT regimen containing fludarabine, cyclophosphamide, and rituximab (FCR). The prognostic implications of the immunoglobulin heavy chain variable gene (IGHV) mutation status have also changed. Unmutated IGHV is associated with inferior PFS and overall survival after FCR and inferior PFS with fixed-duration therapy with venetoclax and anti-CD20 monoclonal antibody but not with continuous BTK inhibitor treatment. Key Messages: (1) Genetic variables (e.g., TP53 aberration, IGHV mutation, complex karyotype) have a prognostic significance in CLL patients treated with targeted therapy. (2) Understanding the prognostic and predictive values of these markers is critical for the development of a risk-adapted treatment strategy in CLL.

Chronic lymphocytic leukemia (CLL) is a disease of malignant B cells whose survival and proliferation are driven by the activation of the B-cell receptor (BCR) signaling pathway and overexpression of pro-survival proteins such as B-cell lymphoma-2 (BCL2). The current standard of care leverages these cellular mechanisms by targeting the BCR signaling pathway with a Bruton’s tyrosine kinase (BTK) inhibitor (BTKi) or promoting cellular apoptosis with a BCL2 inhibitor (BCL2i) [1]. Ibrutinib, acalabrutinib, and zanubrutinib are covalent BTKis approved by the US Food and Drug Administration for the treatment of CLL. Venetoclax is the only BCL2i approved to date which is frequently used in combination with an anti-CD20 monoclonal antibody such as rituximab and obinutuzumab.

Treatment regimens incorporating targeted agents have dramatically improved the patient outcome and fundamentally changed the treatment landscape of CLL from chemoimmunotherapy (CIT) to targeted therapy. In randomized frontline trials comparing BTKi-based regimens to CIT, progression-free survival (PFS) was significantly better with BTKis across the studies [2‒6]. Additionally, overall survival (OS) benefit was observed in the ECOG E1912 study which compared ibrutinib and rituximab (IR) with fludarabine, cyclophosphamide, and rituximab (FCR) in fit, younger patients with treatment-naïve (TN) CLL (3-year OS: 99% for IR and 92% for FCR) [4]. The OS improvement continued to be observed in the recently updated data from the E1912 study at a median follow-up of 5.8 years [7]. Initial treatment with venetoclax plus obinutuzumab (VO) also improved PFS compared to chlorambucil and obinutuzumab among patients with older age or comorbidities [8‒10]. More recently, the CLL13 study comparing venetoclax-based regimens with CIT in fit, younger TN CLL patients demonstrated higher rates of undetectable minimal residual disease (uMRD) and improved PFS in the VO-containing arms [11‒13].

With shifts in treatment approaches and outcomes, changes in the prognostic significance of markers are inevitable. Genetic variables such as complex karyotype (CK) and selected driver gene mutations have gained prognostic importance, while clinical staging and age have become less critical in the context of targeted therapy. Not only the type of treatment but also the treatment duration can affect the role of prognostic markers. For instance, the attainment of uMRD has been associated with prolonged treatment-free survival after fixed-duration therapy with CIT. However, uMRD is uncommonly observed and does not impact long-term outcome during continuous BTKi monotherapy [9, 14, 15]. Critical to the development of a risk-adapted treatment approach in CLL is a precise understanding of the impact of each prognostic marker and its predictive value in selecting patients who might benefit from specific treatment approaches [16]. This review assesses established and emerging prognostic markers in CLL in relevance to targeted therapy.

TP53 gene, mapped at chromosomal band 17p13.1 and encoding p53 tumor suppressor protein, has a critical role in regulating cellular survival signals [17, 18], apoptosis [19, 20], and DNA repair machinery [21]. Most TP53 mutations occur within the DNA binding domain of the gene (97%) [22, 23] and either the deletion of the short arm of chromosome 17 (del17p) or mutation of TP53 has functional consequences as well as concurrent deletion and mutation of the gene [24, 25]. Herein, we refer to del17p and/or TP53 mutation as TP53 aberration.

TP53 aberration is found in 5%–12% of TN CLL patients and up to 30% of relapsed or refractory (RR) CLL [26]. Prospective and retrospective studies demonstrated the strong negative impact of TP53 aberration on CLL outcome, which manifests as primary resistance to or short duration of remission after CIT and inferior survival [27]. In studies investigating CLL driver genes in CIT-treated CLL patients, TP53 mutation had a more negative impact on prognosis than any other mutations (i.e., NOTCH1, SFB1, MYD88, BIRC3) [28], increasing the risk of death by 4-fold [29‒31]. Patients with TP53 aberration have inferior PFS and OS after initial treatment with FCR compared to those with wild-type TP53 (median PFS and OS: 15 and 42 months for the TP53 aberrant group vs. 59 months and not reached for the wild-type TP53 group, respectively) [27]. As a result, International Workshop on CLL (iwCLL) guidelines recommend assessment of TP53 using both FISH and sequencing for all CLL patients before treatment initiation [32].

The outcome of TP53 aberrant CLL markedly improved with targeted therapy (Table 1). In the RESONATE-17 study which exclusively enrolled RR patients with del17p, treatment with ibrutinib resulted in 2-year PFS and OS of 63% and 75%, respectively [33], which contrasted with the historical median PFS of 3 months when FCR was used in this population [34]. Response to a BTKi is particularly durable when it is given as initial therapy. In the NIH phase 2 study including 34 TN CLL patients with TP53 aberration, continuous treatment with single-agent ibrutinib led to 5-year PFS and OS of 70% and 85%, respectively [35]. A pooled analysis of 89 TN, TP53 aberrant CLL patients treated with ibrutinib demonstrated similar results (4-year PFS: 79%, 4-year OS: 88%) [36].

Table 1.

Clinical outcome of CLL patients with or without TP53 aberration

StudyTreatmentN, allN, TP53 aberration*OutcomeRef
TN 
 GCLL8 FCR 316 72 Median PFS: 15 months with TP53MUT versus 59 months without TP53MUT (HR 3.2, p < 0.001); Median OS: 42 months with TP53MUT versus not reached without TP53MUT (HR 4.4, p < 0.001) [27
 NIH TP53 cohort IBR 34 34 (all) 5 years PFS: 70%; 5 years OS: 85% [35
 Pooled analysis of patients with TP53 ab IBR +/− CD20 mAb 89 89 (all) 4 years PFS: 79%; 4 years OS: 88% [36
 Alliance A041202 IBR+/−R 364 34 4 years PFS not reported based on the TP53 status; No PFS or OS difference in subgroups with or without TP53 ab [37
 SEQUOIA Arm D ZANU 109 109 (all) 18 months PFS: 89%; 18 months OS: 95% [38
 CLL14 VO 216 49 5 years PFS: 41% with TP53 ab versus 66% without TP53 ab; 5 years OS: 60% with TP53 ab versus 86% without TP53 ab [10
 MDACC phase 2 IV 80 11 TP53MUT; 14 del17p PFS and OS not reported by the TP53 status; BM-uMRD rate after cycle 24: 45% with TP53MUT, 66% with del17p versus 66% for overall study population [39
 CAPTIVATE IV 164 32 PFS and OS not reported by the TP53 status; Best BM-uMRD rate: 66% with TP53 ab versus 72% without TP53 ab [40
 GLOW IV 106 Outcome data not separately reported by the TP53 status [41
 CLL2-GIVe2 IVO 41 41 (all) 2 years PFS: 95%; 2 years OS: 95% [42
 DFCI AVO 68 41 PFS and OS not reported by the TP53 status; BM-uMRD rate at cycle 16: 83% with TP53 ab versus 89% without TP53 ab [43
 BOVEN ZVO 47 Outcome data not separately reported by the TP53 status [44
RR      
 RESONATE IBR 195 104 Median PFS: 41 months with TP53 ab versus not reached without TP53 ab; Median OS: 62 months with del17p versus 68 months overall [45
 ELEVATE-RR ACA 268 121 Median PFS: 33 months with del17p versus 38 months overall [46
IBR 265 120 Median PFS: 28 months with del17p versus 38 months overall 
 ALPINE ZANU 327 45 2 years PFS: 73% with TP53 ab versus 78% overall [47
IBR 325 50 2 years PFS: 55% with TP53 ab versus 66% overall 
 MURANO VR 194 17 Median PFS: 37 months with TP53 ab versus 57 months without TP53 ab; 5 years OS: 70% with TP53 ab versus 82% overall [48
 CLARITY IV 54 11 Outcome data not separately reported by the TP53 status [49
Mixed: TN and RR      
 Pooled analysis of high risk CLL ACA+/−O 313 TN; 488 RR 64 TN; 219 RR 4 years PFS: 76–77% with TP53 ab TN; 4 years OS: 89% with TP53 ab TN; 3 years PFS: 54% with TP53 ab RR; 3 years OS: 73% TP53 ab RR [50
 OSU IVO 25 TN; 25 RR 3 TN; 1 RR Outcome data not separately reported by the TP53 status [51
StudyTreatmentN, allN, TP53 aberration*OutcomeRef
TN 
 GCLL8 FCR 316 72 Median PFS: 15 months with TP53MUT versus 59 months without TP53MUT (HR 3.2, p < 0.001); Median OS: 42 months with TP53MUT versus not reached without TP53MUT (HR 4.4, p < 0.001) [27
 NIH TP53 cohort IBR 34 34 (all) 5 years PFS: 70%; 5 years OS: 85% [35
 Pooled analysis of patients with TP53 ab IBR +/− CD20 mAb 89 89 (all) 4 years PFS: 79%; 4 years OS: 88% [36
 Alliance A041202 IBR+/−R 364 34 4 years PFS not reported based on the TP53 status; No PFS or OS difference in subgroups with or without TP53 ab [37
 SEQUOIA Arm D ZANU 109 109 (all) 18 months PFS: 89%; 18 months OS: 95% [38
 CLL14 VO 216 49 5 years PFS: 41% with TP53 ab versus 66% without TP53 ab; 5 years OS: 60% with TP53 ab versus 86% without TP53 ab [10
 MDACC phase 2 IV 80 11 TP53MUT; 14 del17p PFS and OS not reported by the TP53 status; BM-uMRD rate after cycle 24: 45% with TP53MUT, 66% with del17p versus 66% for overall study population [39
 CAPTIVATE IV 164 32 PFS and OS not reported by the TP53 status; Best BM-uMRD rate: 66% with TP53 ab versus 72% without TP53 ab [40
 GLOW IV 106 Outcome data not separately reported by the TP53 status [41
 CLL2-GIVe2 IVO 41 41 (all) 2 years PFS: 95%; 2 years OS: 95% [42
 DFCI AVO 68 41 PFS and OS not reported by the TP53 status; BM-uMRD rate at cycle 16: 83% with TP53 ab versus 89% without TP53 ab [43
 BOVEN ZVO 47 Outcome data not separately reported by the TP53 status [44
RR      
 RESONATE IBR 195 104 Median PFS: 41 months with TP53 ab versus not reached without TP53 ab; Median OS: 62 months with del17p versus 68 months overall [45
 ELEVATE-RR ACA 268 121 Median PFS: 33 months with del17p versus 38 months overall [46
IBR 265 120 Median PFS: 28 months with del17p versus 38 months overall 
 ALPINE ZANU 327 45 2 years PFS: 73% with TP53 ab versus 78% overall [47
IBR 325 50 2 years PFS: 55% with TP53 ab versus 66% overall 
 MURANO VR 194 17 Median PFS: 37 months with TP53 ab versus 57 months without TP53 ab; 5 years OS: 70% with TP53 ab versus 82% overall [48
 CLARITY IV 54 11 Outcome data not separately reported by the TP53 status [49
Mixed: TN and RR      
 Pooled analysis of high risk CLL ACA+/−O 313 TN; 488 RR 64 TN; 219 RR 4 years PFS: 76–77% with TP53 ab TN; 4 years OS: 89% with TP53 ab TN; 3 years PFS: 54% with TP53 ab RR; 3 years OS: 73% TP53 ab RR [50
 OSU IVO 25 TN; 25 RR 3 TN; 1 RR Outcome data not separately reported by the TP53 status [51

ab, aberrations; ACA, acalabrutinib; AVO, acalabrutinib, venetoclax, obinutuzumab; BM-uMRD, bone marrow undetectable minimal residual disease; CD20 mAb, anti-CD20 monoclonal antibody; CIT, chemoimmunotherapy; CLL, chronic lymphocytic leukemia; del17p, deletion 17p; FCR, fludarabine, cyclyophosphamide, rituximab; HR, hazard ratio; IBR, ibrutinib; IV, ibrutinib, venetoclax; IVO, ibrutinib, venetoclax, obinutuzumab; M, months; MUT, mutation; N, number; O, obinutuzumab; OS, overall survival; PFS, progression-free survival; R, rituximab; Ref, reference; RR, relapsed or refractory CLL; TN, treatment-naive CLL; VO, venetoclax, obinutuzumab; VR, venetoclax, rituximab; Y, years; ZANU, zanubrutinib; ZVO, zanubrutinib, venetoclax, obinutuzumab.

*TP53 aberration: TP53 mutation or del17p.

Long-term experience with BTKis has provided novel insights relevant to the role of TP53 aberration in CLL. First, TP53 aberration is associated with less durable response to a BTKi in RR CLL but not in TN CLL. The Alliance A041202 study is a three-arm randomized study testing two ibrutinib-containing regimens (ibrutinib with or without rituximab) and conventional CIT with bendamustine and rituximab (BR) in TN CLL [37]. At a median follow-up of 55 months, the study showed no difference in PFS based on the TP53 aberration status within the two ibrutinib-containing arms, suggesting that ibrutinib can overcome the poor prognostic impact of TP53 aberration in the frontline setting. However, TP53 aberration continues to be associated with shorter PFS in RR CLL treated with ibrutinib (median PFS: 36–41 months) [14, 45]. Second, TP53 aberration is associated with increased risks of clonal evolution and subsequent treatment resistance to BTKis. BTK and/or PLCG2 mutations, found in up to 70%–80% of CLL patients who progress on a BTKi [52‒54], occur three times more frequently among patients with TP53 aberration than those without the aberration [55]. Third, the prognostic value of TP53 aberration can change by the type and the allele frequency of the aberration. Approximately 30%–50% of TP53 aberrant CLL presents as monoallelic TP53 aberration (either deletion or mutation), which has been classically associated with inferior response to CIT [56‒58]. A study by Brieghel et al. [59] demonstrated excellent outcomes with ibrutinib across patients with mono- and bi-allelic TP53 aberration. Similarly, low-burden TP53 mutations (variant allele frequency <10–15%) have been associated with poor outcome after CIT [60, 61], but not during treatment with a BTKi [62].

Selective BTKis with more favorable safety profiles than ibrutinib are approved for the treatment of CLL and continue to accumulate data specific to TP53 aberration. Of these, zanubrutinib has been tested in a clinical trial with a large dedicated cohort of TN CLL patients with del17p (n = 109) and achieved excellent initial response (18-month PFS: 89%) [38]. In RR CLL, the efficacy of acalabrutinib in TP53 aberrant CLL was comparable to that of ibrutinib, a finding supported by the 54% 3-year PFS rate in a pooled analysis of 219 patients treated with acalabrutinib [50]. There are two randomized trials directly comparing more versus less selective BTKis in RR CLL, each of which included over 100 patients with TP53 aberration. The ELEVATE-RR study compared acalabrutinib with ibrutinib in RR CLL including 120 patients with del17p, which showed a more favorable safety profile of acalabrutinib and no difference in PFS between the two treatment arms [46]. The ALPINE study comparing zanubrutinib and ibrutinib in RR CLL included 150 patients with TP53 aberration and demonstrated more favorable 2-year PFS in the zanubrutinib arm (73%) than the ibrutinib arm (55%) [47].

The observed differences in the efficacy of BTKis are attributable to the differences in dosing and pharmacokinetic profiles of these agents. The twice-daily (BID) dosing can maintain higher BTK occupancy than the once-daily dosing of acalabrutinib [63] or zanubrutinib [64]. The BID dosing of zanubrutinib achieves 8-fold higher plasma drug exposure than ibrutinib and has a 2- to 4-fold longer half-life than acalabrutinib [65]. In the absence of data providing a head-to-head comparison of zanubrutinib and acalabrutinib, it is difficult to determine which one of the two selective BTKis is better in terms of efficacy and safety. It is also difficult to determine how much the differences in the pharmacokinetic properties of BTKis contribute to the differences in clinical outcomes. Overall, the current data support BTK inhibition as an important treatment strategy for CLL patients with TP53 aberration and provide a strong rationale to use selective BTKis over ibrutinib, given improved tolerance to and more favorable safety profiles of selective BTKis.

Among patients treated with venetoclax-based regimens, TP53 aberration continues to be associated with an inferior prognosis, although the patient outcomes are still markedly better than those with CIT. The CLL14 study is a randomized phase 3 trial comparing 1-year of VO and 6 cycles of chlorambucil and obinutuzumab in TN CLL [66]. At the median follow-up of 65 months, the 5-year PFS of the VO arm was 41% for a subgroup with TP53 aberration and 66% for those without TP53 aberration, raising concerns for the limited durability of remission in TP53 aberrant CLL treated with fixed-duration therapy [10]. Treatment of RR CLL patients with a 2-year duration of venetoclax plus rituximab (VR) in the MURANO study also showed that the remission is less durable (4-year PFS: 33%) and less deep (uMRD in peripheral blood [PB] at the end of treatment: 36%) in the del17p subgroup compared to the overall study population (4-year PFS: 57%, PB uMRD at the end of treatment: 62%) [67]. Although the 5-year PFS of 41% on VO is seemingly lower than up to 70% 5-year PFS observed on a continuous BTKi, one should not directly compare the efficacy of these approaches without prospective randomized data. Current NCCN guidelines recommend both time-limited venetoclax plus anti-CD20 monoclonal antibody and continuous BTKi therapy as preferred regimens in CLL.

To overcome the limitation of established regimens, several studies are testing targeted combinations incorporating a BTKi and venetoclax with or without anti-CD20 monoclonal antibody coupled with MRD-guided treatment cessation. Several of these trials were enriched with TP53 aberrant CLL. The CLL2-GIVe study exclusively enrolled 41 TN patients with TP53 aberrant CLL and reported high rates of complete response (59%) and bone marrow (BM) uMRD (66%) at the end of combination therapy with ibrutinib plus VO (IVO) [42]. Two additional studies tested targeted triplet regimens replacing ibrutinib with a selective BTKi acalabrutinib (AVO) [68] or zanubrutinib (ZVO) [44]. Of these, AVO achieved high rates of BM uMRD in subgroups with (83%) or without TP53 aberration (89%). The study testing ZVO had only 2 patients with TP53 aberration. Studies testing the doublet regimen of ibrutinib plus venetoclax (IV) had relatively small numbers of patients with TP53 aberration, limiting one from drawing strong conclusions from these studies specific to the TP53 subset. Despite the limitation, initial responses to IV were comparable between the overall study population and those with TP53 aberration in several trials. For example, BM uMRD rate was 66% in the TP53 aberrant subgroup and 68% in the overall study population of the CAPTIVATE study [40]. Other studies with a smaller number of patients with TP53 aberration (n = 7–18) reported BM uMRD rates ranging between 56% and 64% in TN CLL [39, 41] and 36% in RR CLL [49].

Outcome with a sequential combination of targeted therapy has been reported in two studies. In the study by Thompson et al. [69], ibrutinib-treated CLL patients with selected high-risk markers received venetoclax as an adjunct to ibrutinib (I + V). Over half of the patients in this study had TP53 aberration (28 of 45 patients) and the rate of BM uMRD after 12 cycles of I + V was 57%. Another study by Scarfo et al. [70] tested the addition of ibrutinib to venetoclax in RR CLL patients who had detectable MRD after 12 cycles of initial treatment with venetoclax (V + I). Despite all patients in this study being previously treated with CIT and a third of the study population having TP53 aberration (11 of 38 patients), 84% achieved uMRD in both PB and BM after a median of 7 months of V + I. Deep responses can be achieved with the addition of a PI3K inhibitor, umbralisib, and anti-CD20 antibody, ublituximab, to ibrutinib (77% PB uMRD at best response); however, this study included only 2 patients with del17p, and the FDA has withdrawn approval for umbralisib due to safety concerns [71].

The key lesson learned from the recent studies in CLL is that the combination of targeted agents increases the likelihood of uMRD in high-risk CLL, and the MRD-driven treatment cessation is feasible for patients with TP53 aberration. In a pooled analysis of 51 patients with TP53 aberration treated with ibrutinib or venetoclax plus anti-CD20 monoclonal antibody regimens across 3 trials, a third of the patients were able to stop therapy after achieving uMRD and had median PFS of 2 years after MRD-guided treatment cessation [72]. Further studies are needed to determine the duration of deep remission in TP53 aberrant CLL, the best regimen that can provide durable disease control in this subgroup, and its relevance to treatment-free survival and OS.

CK is defined by the detection of 3 or more chromosomal abnormalities in the metaphase cytogenetics performed with cytokine or CpG oligonucleotide stimulation. Some of the abnormalities that are well-known and relevant to the prognosis of CLL include deletion of 6q, 11q, 13q, and 17p, and trisomy 12 [73], which can be sensitively detected by interphase FISH. Cytogenetics can help identify copy number changes and chromosomal rearrangements affecting regions not covered by the conventional CLL FISH panel. In a large retrospective study including 5,290 CLL patients, CK (≥3 abnormalities) was found in 15% of the patients and was associated with shorter OS [74]. Intriguingly, not all patients with CK had an adverse outcome in this study. Approximately 10% of the patients with CK had trisomies commonly affecting chromosome 12 or 19 and had excellent survival. Further, the study revealed highly CK with ≥5 abnormalities had a stronger negative impact on survival than ≥3 abnormalities.

The prognostic relevance of CK in 456 BTKi-treated patients has been reported by the group at Ohio State University (OSU) [75, 76]. This study demonstrated that CK was a continuous variable that increased the risk of progression or death on ibrutinib by 7% with each additional abnormality. The OS difference was more pronounced in subgroups with ≥5 abnormalities compared to ≥3. Another study by Thompson et al. [77] also demonstrated inferior event-free survival and OS associated with CK (≥3 abnormalities) in 88 RR patients treated with ibrutinib-based regimens (median EFS and OS: 19 and 25 months for the CK group vs. 39 months and not reached for the non-CK group, respectively). It is important to note that most (78%) of the patients in the OSU study and all patients in the study by Thompson et al. [78] had RR CLL at the time of ibrutinib initiation. Smaller data exist for the prognostic role of CK in TN CLL patients treated with a BTKi. In the GIMEMA LLC1114 study including 98 TN CLL patients treated with ibrutinib, CK (≥3 abnormalities) was associated with inferior PFS.

CK has been investigated in TN and RR patients treated with VO-containing regimens. The CLL13 study was a four-arm trial randomizing over 900 TN patients to CIT, VR, VO, or IVO, which reported that CK ≥5 abnormalities were associated with significantly shorter PFS across all treatment arms [79, 80]. Three or four abnormalities resulted in a shorter PFS with CIT (HR 2.49, p = 0.003), but not with the venetoclax-based treatment arms. Interestingly, 8q and 18q translocations showed significantly inferior PFS, providing data for the first time on specific chromosomal regions of clinical significance. The CLL14 study led to distinct conclusions on the role of CK in TN CLL patients treated with VO [81]. The CK subgroup with ≥3 abnormalities had a trend toward inferior PFS and OS without reaching statistical significance (2-year PFS and OS: 79% and 88% with CK vs. 91% and 93% without CK). Only 10 patients in this study had high CK (≥5), and the outcomes were not significantly different in this subgroup. In RR CLL, CK was assessed using array comparative genomic hybridization (sCGH) in the MURANO study [48]. Patients with ≥3 abnormalities by aCGH had markedly inferior PFS (42 months) than those without (60 months), highlighting the prognostic importance of CK in RR CLL patients treated with VR. A smaller study using conventional cytogenetics also reported a higher risk of disease progression associated with CK (≥3) in RR CLL (HR 6.6, p = 0.005) [82].

CK is not as frequently tested as other prognostic markers in CLL, which limits further investigation and incorporation of this marker in novel prognostic models [83]. Data are sparse for the biological implication and the prognostic role of individual chromosomal loci beyond those commonly detected by FISH. Improved utilization of cytogenetics in clinical practice, systematic analyses of large clinical datasets, and reliable laboratory models representative of karyotypic complexity are needed to further our understanding of CK in CLL.

Somatic hypermutation of the immunoglobulin heavy chain variable region gene (IGHV) occurs during affinity maturation of B cells at the germinal center [84]. In CLL, IGHV mutation dichotomizes CLL into two distinct biological and clinical phenotypes. Mutated IGHV (M-IGHV), defined by ≥2% divergence from the consensus germline sequence, is characterized by high-affinity binding of the BCR, a more indolent clinical course, and more durable remission after CIT [85, 86]. Treatment with FCR, for instance, can achieve long-lasting remission in young and fit patients with M-IGHV (12.8-year PFS: 54% with M-IGHV, 9% with unmutated IGHV [U-IGHV]) [87, 88].

Intriguingly, a group of patients with IGHV3-21 usage breaks the conventional IGHV classification. While most of these patients are categorized as M-IGHV, their clinical course mimics that of those with U-IGHV [89]. Nearly half of these patients have a stereotyped pattern of immunoglobulin gene usage called subset 2, which is defined by the presence of IGHV3-21 and additional characteristics of IGHV including short CDR3 sequence (9 amino acids) and acidic amino acid in the landmark location (most frequently aspartate) [90]. Recent studies demonstrated a high incidence of point mutations affecting the immunoglobulin light chain gene IGLV3-21R110 in subset 2, which induces autonomous BCR signaling through the interaction of neighboring BCRs [91, 92]. While subset 2 and IGLV3-21R110 mutation define a unique biological subgroup [74], detailed testing of the IGHV stereotype and the light chain gene sequences has not been widely adopted in clinical practice, and the prognostic role of these markers in the context of targeted therapy remains unclear.

The prognostic value of IGHV mutation has changed with targeted therapy with notable differences in the durability of response to continuous and time-limited treatments. Continuous BTKis lead to high rates of initial responses and durable disease control irrespective of IGHV status [5, 93]. In contrast, fixed-duration venetoclax-based therapy has shown marked differences among IGHV subgroups in the duration of treatment-free remission and PFS after treatment cessation [8]. Median PFS was approximately 5 years for TN patients with U-IGHV after VO therapy and not reached for those with M-IGHV in the CLL14 study [66].

Trials testing targeted combinations identified notable differences in MRD kinetics by IGHV status. Patients with U-IGHV are highly sensitive to IV doublet therapy, leading to faster attainment and higher rates of uMRD than those with M-IGHV [40, 94]. However, the uMRD in patients with U-IGHV was not durable, and MRD conversion from undetectable to detectable disease occurred at a rate of approximately 10% per year for the U-IGHV subgroup after stopping the IV combination [95]. Overall, IGHV mutation status is an important prognostic marker associated with remission duration and MRD kinetics among patients receiving time-limited targeted therapy.

uMRD is defined by <1 CLL cell per 10,000 leukocytes (<10−4) based on multicolor flow cytometry, real-time quantitative polymerase chain reaction (PCR), or NGS-based IGH sequencing [96]. Multicolor flow cytometry is a widely available method with sensitivity limited to 10−4. The NGS method is highly sensitive (10−6) but requires a baseline sample to identify disease-specific IGHV sequences prior to MRD testing. The PCR method is also sensitive (10−5) but labor-intensive due to its requirement for patient-specific primers. Initial evidence for MRD as a post-treatment prognostic marker was generated in the context of CIT. Nearly half of the TN patients treated with FCR achieved BM uMRD and those who attained uMRD had significantly longer PFS and OS than patients with detectable disease (shown in Table 2) [97, 98].

Table 2.

Rates of undetected minimal residual disease in bone marrow

StudyTreatmentPopulationMRD testing method and timepointIGHV statusBM uMRD unless otherwise specifiedRef
CIT CLL8 FCR 210 TN FC at EOT M-IGHV 50% [97
U-IGHV 41% 
MDACC FCR300 FCR 300 TN FC at EOT M-IGHV 51% [88
U-IGHV 33% 
Continuous BTKi NIH phase 2 IBR 53 TN + 33 RR FC, best MRD M-IGHV 10% [14
U-IGHV 4% 
iLLUMINATE IBR-O 113 TN FC, best MRD Overall study population 25% [99
BCL2i + anti-CD20 mAb CLL14 VO 216 TN ASO-PCR at EOT M-IGHV 74% (PB uMRD) [66
U-IGHV 79% (PB uMRD) 
MURANO VR 194 RR FC at EOT (24C) M-IGHV 43% (PB uMRD) [48
U-IGHV 46% (PB uMRD) 
BTKi + BCL2i doublet MDACC phase 2 IV 80 TN FC at EOCT (24C) M-IGHV 67% [39
U-IGHV 54% 
CAPTIVATE IV 159 TN FC, best MRD M-IGHV 53% [40
U-IGHV 64% 
GLOW IV 106 TN NGS at EOCT (15C) M-IGHV 41% [41
U-IGHV 60% 
CLARITY IV 54 RR FC at EOCT (24 months) Overall study population 36% [49
Scarfo et al. IBR added to V 38 RR FC at EOCT (12C) Overall study population 84% (both PB and BM uMRD) [70
BTKi + BCL2i + anti-CD20 mAb triplet CLL2-GIVe IVO 41 TN FC at EOT (15C) Overall study population 66% [42
DFCI phase 2 AVO 37 TN FC at EOCT (24C) Overall study population 84% [68
BOVEN ZVO 39 TN FC, best MRD Overall study population 89% [44
OSU IVO 25 TN + 25 RR FC at EOCT (14C) Overall study population 67% in TN, 50% in RR [51
StudyTreatmentPopulationMRD testing method and timepointIGHV statusBM uMRD unless otherwise specifiedRef
CIT CLL8 FCR 210 TN FC at EOT M-IGHV 50% [97
U-IGHV 41% 
MDACC FCR300 FCR 300 TN FC at EOT M-IGHV 51% [88
U-IGHV 33% 
Continuous BTKi NIH phase 2 IBR 53 TN + 33 RR FC, best MRD M-IGHV 10% [14
U-IGHV 4% 
iLLUMINATE IBR-O 113 TN FC, best MRD Overall study population 25% [99
BCL2i + anti-CD20 mAb CLL14 VO 216 TN ASO-PCR at EOT M-IGHV 74% (PB uMRD) [66
U-IGHV 79% (PB uMRD) 
MURANO VR 194 RR FC at EOT (24C) M-IGHV 43% (PB uMRD) [48
U-IGHV 46% (PB uMRD) 
BTKi + BCL2i doublet MDACC phase 2 IV 80 TN FC at EOCT (24C) M-IGHV 67% [39
U-IGHV 54% 
CAPTIVATE IV 159 TN FC, best MRD M-IGHV 53% [40
U-IGHV 64% 
GLOW IV 106 TN NGS at EOCT (15C) M-IGHV 41% [41
U-IGHV 60% 
CLARITY IV 54 RR FC at EOCT (24 months) Overall study population 36% [49
Scarfo et al. IBR added to V 38 RR FC at EOCT (12C) Overall study population 84% (both PB and BM uMRD) [70
BTKi + BCL2i + anti-CD20 mAb triplet CLL2-GIVe IVO 41 TN FC at EOT (15C) Overall study population 66% [42
DFCI phase 2 AVO 37 TN FC at EOCT (24C) Overall study population 84% [68
BOVEN ZVO 39 TN FC, best MRD Overall study population 89% [44
OSU IVO 25 TN + 25 RR FC at EOCT (14C) Overall study population 67% in TN, 50% in RR [51

ASO-PCR, allele-specific oligonucleotide polymerase chain reaction; AVO, acalabrutinib, venetoclax, obinutuzumab; BCL2i, B-cell lymphoma-2 inhibitor; BM, bone marrow; BTKi, Bruton’s tyrosine kinase inhibitor; C, cycles; CIT, chemoimmunotherapy; CLL, chronic lymphocytic leukemia; EOCT, end of combination therapy; EOT, end of therapy; FC, flow cytometry; FCR, fludarabine, cyclophosphamide, rituximab; IBR, ibrutinib; IGHV, immunoglobulin heavy chain variable gene; IV, ibrutinib, venetoclax; IVO, ibrutinib, venetoclax, obinutuzumab; M: months; mAb, monoclonal antibody; M-IGHV, mutated IGHV; MRD, minimal residual disease; NGS, next generation sequencing; O, obinutuzumab; PB, peripheral blood; Ref, reference; RR, relapsed or refractory CLL; TN, treatment-naive CLL; U-IGHV, unmutated IGHV; uMRD. undetectable minimal residual disease; V, venetoclax; VO, venetoclax, obinutuzumab; VR, venetoclax, rituximab; ZVO, zanubrutinib, venetoclax, obinutuzumab.

Unlike CIT, uMRD is uncommonly observed among patients on BTKi monotherapy. Attainment of low- (<10−2) versus high-level (≥10−2) MRD was not associated with differences in PFS during BTKi therapy [14]. The addition of an anti-CD20 monoclonal antibody [3, 5] or CIT [100‒102] to a BTKi can improve the uMRD rate. In the ELEVATE-TN study, the addition of obinutuzumab to acalabrutinib improved 4-year PFS by nearly 10% and the rate of PB uMRD by 28% [103]. Initial treatment with venetoclax-based regimens substantially increases the rate of PB uMRD to 76% (PCR method) [10]. In the CLL14 study, attainment of uMRD at the end of fixed-duration VO therapy was associated with higher PFS and OS compared to patients with detectable MRD (4-year post-treatment PFS and OS: 77% and 89% with uMRD vs. 36% and 64% without uMRD, respectively). Similar to the TN population, RR patients treated who achieved uMRD after treatment with VR had superior OS than those with detectable disease (3-year post-treatment OS: 95% with uMRD vs. 73% without uMRD) [48]. Taken together, post-treatment uMRD predicts durable remission and improved survival in patients treated with fixed-duration therapy containing venetoclax and anti-CD20 antibody.

Targeted combinations can achieve high rates of uMRD as well (Table 2). In TN CLL treated with IV doublet, approximately half of the patients achieved BM uMRD and the uMRD rate was higher in the U-IGHV subgroup (56–77%) than the M-IGHV subgroup (∼50%) [39, 40]. The BM uMRD rate in RR CLL treated with IV is over 10% lower than that observed in the TN population (36% in RR) [49]. Triplet combination can further increase the BM uMRD rate in TN CLL with reported rates at 66–78% with IVO [42, 51], 78–89% with AVO [68], and 89% with ZVO [44]. Extended follow-up and serial assessment of the MRD status are needed to better characterize uMRD as a post-treatment prognostic marker in the setting of targeted combination therapy.

Integration of Prognostic Markers

CLL International Prognostic Index (CLL-IPI) is a widely used, weighted scoring system integrating clinical (age, clinical stage), molecular (IGHV, TP53 aberration status), and serologic markers (beta-2 microglobulin [B2M]) for risk stratification of previously untreated CLL patients [31]. While CLL-IPI was developed in the era of CIT and its survival data are irrelevant to current practice, it remains to be a useful tool to predict time to first therapy since the criteria for treatment initiation have not changed in CLL.

Several studies reported novel prognostic tools more relevant to targeted therapy. CLL-BALL utilizes B2M, lactate dehydrogenase (LDH), hemoglobin, and time from the last treatment to stratify patients into groups with distinct OS [104]. However, this model did not test PFS as an endpoint and patients in the discovery dataset received heterogeneous treatments including ibrutinib, idelalisib, and CIT. To overcome these limitations, CLL4 has been developed from over 800 patients treated with ibrutinib [83]. This model selected four factors: TP53 aberration status, prior treatment status, B2M, and LDH, based on machine-learning algorithms and conventional multivariable analysis. The high-risk group defined by CLL4 had the shortest PFS and OS and the highest incidence of BTK/PLCG2 mutations as well as Richter’s transformation. The study also has limitations related to the lack of integration of CK and the inclusion of prior treatment status as a factor that can limit its applicability to patients receiving BTKis as initial therapy. Further research is needed to integrate novel molecular markers in the risk stratification of CLL and develop prognostic tools relevant to venetoclax-based therapy.

CD49d

CD49d is a surface molecule of the integrin heterodimer α4 subunit which mediates leukocyte adhesion via very late antigen 4 (VLA-4). It is critical for leukocyte interaction through inside-out activation triggered by the BCR signaling pathway [105, 106], and its expression promotes the microenvironment-mediated proliferation of CLL cells, leading to a poor prognosis [107]. CD49d expression is assessed by flow cytometry, with >30% expression set as a cutoff for positivity [108]. In a study of 2,972 CLL patients conducted in the era of CIT, CD49d positivity was associated with inferior survival (10-year OS: 62% for CD49+ CLL vs. 84% for CD49d− CLL) [109]. Smaller data exist for ibrutinib-treated CLL patients which also demonstrated inferior PFS associated with CD49d positivity (HR 3.32, p = 0.009) [108]. There are no data on the prognostic role of CD49d in patients treated with venetoclax-based therapy.

Deletion 11q and/or ATM Mutation

Inactivation of ataxia telangiectasia mutated (ATM) gene with mutation or deletion of chromosome 11q (del11q) decreases response to DNA double-strand breaks and increases the risk of lymphoid malignancies [110]. While patients with del11q or ATM mutations had poor prognoses on CIT [111‒113], targeted agents dramatically improved the outcome of this population, leading to PFS and OS comparable to [48, 93] or even longer than those without del11q [114]. Del11q and ATM mutation are no longer considered adverse prognostic factors for patients receiving targeted therapy.

Serologic Markers

Serologic markers of disease activity in CLL include B2M and LDH. Elevated B2M and LDH at pre-treatment are associated with poor prognosis in CLL treated with CIT and targeted therapy including BTKi- and venetoclax-based regimens [31, 83, 104, 115]. The serologic markers dynamically change over time and with therapy. To our knowledge, no studies have investigated the prognostic value of serial monitoring of serologic markers.

Patient-Specific Factors

Patient-specific factors, particularly age and comorbidities, can predict risks associated with CLL therapy and assist in individualized treatment decisions. Age at diagnosis has long been recognized as an independent predictor of survival in CLL, with younger age being associated with a more aggressive disease course and shorter life expectancy compared to the age-matched general population [116, 117]. Moreover, selected comorbidities affecting vascular, upper gastrointestinal, and/or endocrine systems are linked to decreased OS and shorter time to treatment in large datasets of over 4,000 patients with CLL [118, 119]. While older patients may have relatively indolent disease courses, the presence of an increased burden of comorbidity has been shown to significantly impair tolerance to disease- and treatment-related complications and decrease survival [120]. However, these findings were generated when CIT was the prevailing treatment choice and may be less relevant during targeted therapy. Several trials have specifically demonstrated safety and excellent tolerance to the established targeted therapy regimens in older adults or those with comorbidities. For instance, the Alliance A041202 study enrolled patients with age 65 years or older and showed lower incidences of high-grade adverse events with ibrutinib compared to BR. The CLL14 study also enrolled relatively older patients (median age 72 years) with a cumulative illness rating scale (CIRS) score greater than 6 or impaired renal function (creatinine clearance <70 mL/min) and demonstrated VO was well tolerated in this population [10]. Based on these data, patients with older age and comorbidities should be considered for the current standard of care therapy with a BTKi or VO.

Additional patient-specific factors most relevant to the selection of targeted therapy include history of cardiovascular disease, history or increased risk of serious bleeding, and drug-drug interaction [1]. Because BTKis are associated with hypertension, arrhythmia, and sudden cardiac deaths, patients with a prior history of cardiac disease or uncontrolled hypertension are generally recommended to avoid this therapeutic class or at least use a selective BTKi [121‒123]. Bleeding is a common side effect of all BTKis that occurs in similar frequencies in selective and less selective BTKis. Although grade 3 or higher bleeding on a BTKi is rare (<5%), patients with a history of serious bleeding events (i.e., intracranial hemorrhage, large hematoma) or those on coumadin should avoid BTKis [124]. Drug-drug interaction is relevant to all targeted therapy and providers should carefully review concurrent medications. Both BTKis and venetoclax require dose reduction during the use of moderate CYP3A inhibitors and dose interruption when strong CYP3A inhibitors or inducers are used. Venetoclax also requires 50% dose reduction during treatment with a P-gp inhibitor [1].

The introduction of targeted therapy for the treatment of CLL has generated new questions related to the present value of prognostic markers in this disease (shown in Fig. 1). Initial treatment with a BTKi can overcome the poor prognostic value of TP53 aberration in TN CLL. TP53 aberration continues to be a negative prognostic factor for RR CLL patients receiving BTKi therapy and TN and RR patients treated with venetoclax-based, time-limited therapy. CK is linked with unfavorable outcomes across targeted agents. The prognostic value of CK becomes stronger when a higher cutoff of 5 or more abnormalities is used instead of the conventional cutoff of 3. IGHV mutation remains an important prognostic marker predictive of the durability of remission after time-limited targeted therapy, but not with continuous BTKi treatment. These data align with the current iwCLL guidelines, which recommend testing of IGHV mutation status, FISH, and TP53 sequencing as mandatory tests at pre-treatment and karyotyping as a desirable test in the setting of clinical trials [32]. The NCCN guidelines also recognize these tests and list CK as a well-defined prognostic marker (CLL, version 2.2023). To develop an individualized, genomically driven treatment strategy in CLL, further research is needed to validate prognostic markers in predicting treatment response and disease progression, to create risk-adapted strategies that incorporate predictive markers for patient and treatment selection, and to develop reproducible and widely applicable biomarkers for clinical use.

Fig. 1.

Impact of prognostic factors by treatment regimens. Summarizes how the prognostic value of TP53 aberration, CK, immunoglobulin heavy chain variable region gene mutation status, and minimal residual disease changed with targeted therapies. TP53 aberration refers to deletion 17p and/or TP53 mutation. BTKi, Bruton’s tyrosine kinase inhibitor; IGHV, immunoglobulin heavy chain variable gene; mAb, anti-CD20 monoclonal antibody; MRD, minimal residual disease; RR, relapsed or refractory CLL; TN, treatment-naive CLL; VEN, venetoclax.

Fig. 1.

Impact of prognostic factors by treatment regimens. Summarizes how the prognostic value of TP53 aberration, CK, immunoglobulin heavy chain variable region gene mutation status, and minimal residual disease changed with targeted therapies. TP53 aberration refers to deletion 17p and/or TP53 mutation. BTKi, Bruton’s tyrosine kinase inhibitor; IGHV, immunoglobulin heavy chain variable gene; mAb, anti-CD20 monoclonal antibody; MRD, minimal residual disease; RR, relapsed or refractory CLL; TN, treatment-naive CLL; VEN, venetoclax.

Close modal

S.K.: no conflict of interest. I.E.A.: consulting – BeiGene.

I.E.A. acknowledges support from the Leukemia and Lymphoma Society Scholar in Clinical Research Award and the Mayer Foundation.

S.K. and I.E.A. reviewed the literature and wrote the manuscript.

1.
Ahn IE, Brown JR. Selecting initial therapy in CLL. Hematology Am Soc Hematol Educ Program. 2022 Dec 9;2022(1):323–8.
2.
Woyach JA, Ruppert AS, Heerema NA, Zhao W, Booth AM, Ding W, et al. Ibrutinib regimens versus chemoimmunotherapy in older patients with untreated CLL. N Engl J Med. 2018 Dec 27;379(26):2517–28.
3.
Moreno C, Greil R, Demirkan F, Tedeschi A, Anz B, Larratt L, 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.
4.
Shanafelt TD, Wang XV, Kay NE, Hanson CA, O’Brien S, Barrientos J, et al. Ibrutinib-rituximab or chemoimmunotherapy for chronic lymphocytic leukemia. N Engl J Med. 2019 Aug 1;381(5):432–43.
5.
Sharman JP, Egyed M, Jurczak W, Skarbnik A, Pagel JM, Flinn IW, et al. Acalabrutinib with or without obinutuzumab versus chlorambucil and obinutuzumab for treatment-naive chronic lymphocytic leukaemia (ELEVATE-TN): a randomised, controlled, phase 3 trial. Lancet. 2020 Apr 18;395(10232):1278–91.
6.
Tam CS, Brown JR, Kahl BS, Ghia P, Giannopoulos K, Jurczak W, et al. Zanubrutinib versus bendamustine and rituximab in untreated chronic lymphocytic leukaemia and small lymphocytic lymphoma (SEQUOIA): a randomised, controlled, phase 3 trial. Lancet Oncol. 2022 Aug;23(8):1031–43.
7.
Shanafelt TD, Wang XV, Hanson CA, Paietta EM, O’Brien S, Barrientos J, et al. Long-term outcomes for ibrutinib-rituximab and chemoimmunotherapy in CLL: updated results of the E1912 trial. Blood. 2022 Jul 14;140(2):112–20.
8.
Fischer K, Al-Sawaf O, Bahlo J, Fink AM, Tandon M, Dixon M, et al. Venetoclax and obinutuzumab in patients with CLL and coexisting conditions. N Engl J Med. 2019 Jun 6;380(23):2225–36.
9.
Al-Sawaf O, Zhang C, Tandon M, Sinha A, Fink AM, Robrecht S, 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 Sep;21(9):1188–200.
10.
Al-Sawaf O, Zhang C, Jin HY, Robrecht S, Choi Y, Balasubramanian S, et al. Transcriptomic profiles and 5-year results from the randomized CLL14 study of venetoclax plus obinutuzumab versus chlorambucil plus obinutuzumab in chronic lymphocytic leukemia. Nat Commun. 2023 Apr 18;14(1):2147.
11.
Eichhorst B, Niemann C, Kater AP, Fürstenau M, Von Tresckow J, Zhang C, et al. A randomized phase III study of venetoclax-based time-limited combination treatments (RVe, GVe, GIVe) vs standard chemoimmunotherapy (CIT: FCR/BR) in frontline chronic lymphocytic leukemia (CLL) of fit patients: first Co-primary endpoint analysis of the international intergroup GAIA (CLL13) trial. Blood. 2021;138(Supplement 1):71.
12.
Eichhorst B, Niemann C, Aea K. Time-limited venetoclax-obinutuzumab +/− ibrutinib is highly effective and safe in patients with previously untreated chronic lymphocytic leukemia (CLL): final analysis of the CLL14 trial. In: 27th congress of the European hematology association (EHA), Virtual edition; 2022.
13.
Eichhorst B, Niemann CU, Kater AP, Fürstenau M, von Tresckow J, Zhang C, et al. First-Line venetoclax combinations in chronic lymphocytic leukemia. N Engl J Med. 2023 May 11;388(19):1739–54.
14.
Ahn IE, Farooqui MZH, Tian X, Valdez J, Sun C, Soto S, et al. Depth and durability of response to ibrutinib in CLL: 5-year follow-up of a phase 2 study. Blood. 2018 May 24;131(21):2357–66.
15.
Seymour JF. CLL12: a positive answer to a poorly phrased question. Blood. 2022 Jan 13;139(2):151–2.
16.
Stilgenbauer S. Four-factor score for outcome of ibrutinib treatment in chronic lymphocytic leukemia: prognostic model for risk group definition. J Clin Oncol. 2021 Feb 20;39(6):551–3.
17.
Brugarolas J, Chandrasekaran C, Gordon JI, Beach D, Jacks T, Hannon GJ. Radiation-induced cell cycle arrest compromised by p21 deficiency. Nature. 1995 Oct 12;377(6549):552–7.
18.
Mihara M, Erster S, Zaika A, Petrenko O, Chittenden T, Pancoska P, et al. p53 has a direct apoptogenic role at the mitochondria. Mol Cell. 2003 Mar;11(3):577–90.
19.
Villunger A, Michalak EM, Coultas L, Müllauer F, Böck G, Ausserlechner MJ, et al. p53- and drug-induced apoptotic responses mediated by BH3-only proteins puma and noxa. Science. 2003 Nov 7;302(5647):1036–8.
20.
Chipuk JE, Kuwana T, Bouchier-Hayes L, Droin NM, Newmeyer DD, Schuler M, et al. Direct activation of Bax by p53 mediates mitochondrial membrane permeabilization and apoptosis. Science. 2004 Feb 13;303(5660):1010–4.
21.
Gao Y, Ferguson DO, Xie W, Manis JP, Sekiguchi J, Frank KM, et al. Interplay of p53 and DNA-repair protein XRCC4 in tumorigenesis, genomic stability and development. Nature. 2000 Apr 20;404(6780):897–900.
22.
Kato S, Han SY, Liu W, Otsuka K, Shibata H, Kanamaru R, et al. Understanding the function-structure and function-mutation relationships of p53 tumor suppressor protein by high-resolution missense mutation analysis. Proc Natl Acad Sci U S A. 2003 Jul 8;100(14):8424–9.
23.
Scian MJ, Stagliano KE, Ellis MA, Hassan S, Bowman M, Miles MF, et al. Modulation of gene expression by tumor-derived p53 mutants. Cancer Res. 2004 Oct 15;64(20):7447–54.
24.
Oren M, Rotter V. Mutant p53 gain-of-function in cancer. Cold Spring Harb Perspect Biol. 2010 Feb;2(2):a001107.
25.
Athanasakis E, Melloni E, Rigolin GM, Agnoletto C, Voltan R, Vozzi D, et al. The p53 transcriptional pathway is preserved in ATMmutated and NOTCH1mutated chronic lymphocytic leukemias. Oncotarget. 2014 Dec 30;5(24):12635–45.
26.
Stilgenbauer S, Zenz T, Winkler D, Bühler A, Schlenk RF, Groner S, et al. Subcutaneous alemtuzumab in fludarabine-refractory chronic lymphocytic leukemia: clinical results and prognostic marker analyses from the CLL2H study of the German Chronic Lymphocytic Leukemia Study Group. J Clin Oncol. 2009 Aug 20;27(24):3994–4001.
27.
Stilgenbauer S, Schnaiter A, Paschka P, Zenz T, Rossi M, Döhner K, et al. Gene mutations and treatment outcome in chronic lymphocytic leukemia: results from the CLL8 trial. Blood. 2014 May 22;123(21):3247–54.
28.
Rossi D, Rasi S, Spina V, Bruscaggin A, Monti S, Ciardullo C, et al. Integrated mutational and cytogenetic analysis identifies new prognostic subgroups in chronic lymphocytic leukemia. Blood. 2013 Feb 21;121(8):1403–12.
29.
Knight SJ, Yau C, Clifford R, Timbs AT, Sadighi Akha E, Dréau HM, et al. Quantification of subclonal distributions of recurrent genomic aberrations in paired pre-treatment and relapse samples from patients with B-cell chronic lymphocytic leukemia. Leukemia. 2012 Jul;26(7):1564–75.
30.
Guièze R, Robbe P, Clifford R, de Guibert S, Pereira B, Timbs A, et al. Presence of multiple recurrent mutations confers poor trial outcome of relapsed/refractory CLL. Blood. 2015 Oct 29;126(18):2110–7.
31.
International CLL-IPI working group. An international prognostic index for patients with chronic lymphocytic leukaemia (CLL-IPI): a meta-analysis of individual patient data. Lancet Oncol. 2016;17(6):779–90.
32.
Hallek M, Cheson BD, Catovsky D, Caligaris-Cappio F, Dighiero G, Döhner H, et al. iwCLL guidelines for diagnosis, indications for treatment, response assessment, and supportive management of CLL. Blood. 2018 Jun 21;131(25):2745–60.
33.
O’Brien S, Jones JA, Coutre SE, Mato AR, Hillmen P, Tam C, et al. Ibrutinib for patients with relapsed or refractory chronic lymphocytic leukaemia with 17p deletion (RESONATE-17): a phase 2, open-label, multicentre study. Lancet Oncol. 2016 Oct;17(10):1409–18.
34.
Badoux XC, Keating MJ, Wang X, O’Brien SM, Ferrajoli A, Faderl S, et al. Cyclophosphamide, fludarabine, alemtuzumab, and rituximab as salvage therapy for heavily pretreated patients with chronic lymphocytic leukemia. Blood. 2011 Aug 25;118(8):2085–93.
35.
Ahn IE, Tian X, Wiestner A. Ibrutinib for chronic lymphocytic leukemia with TP53 alterations. N Engl J Med. 2020;383(5):498–500.
36.
Allan JN, Shanafelt T, Wiestner A, Moreno C, O’Brien SM, Li J, et al. Long-term efficacy of first-line ibrutinib treatment for chronic lymphocytic leukaemia in patients with TP53 aberrations: a pooled analysis from four clinical trials. Br J Haematol. 2022 Feb;196(4):947–53.
37.
Woyach J, Ruppert A, Heerema N, Zhao W, Booth A, Ding W, et al. Long-term results of alliance A041202 show continued advantage of ibrutinib-based regimens compared with bendamustine plus rituximab (BR) chemoimmunotherapy. Blood. 2021 11/05;138(Supplement 1):639.
38.
Tam CS, Robak T, Ghia P, Kahl BS, Walker P, Janowski W, et al. Zanubrutinib monotherapy for patients with treatment naïve chronic lymphocytic leukemia and 17p deletion. Haematologica. 2020 Oct 13;106(9):2354–63.
39.
Jain N, Keating M, Thompson P, Ferrajoli A, Burger JA, Borthakur G, et al. Ibrutinib plus venetoclax for first-line treatment of chronic lymphocytic leukemia: a nonrandomized phase 2 trial. JAMA Oncol. 2021 Aug 1;7(8):1213–9.
40.
Wierda WG, Allan JN, Siddiqi T, Kipps TJ, Opat S, Tedeschi A, 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 Dec 1;39(34):3853–65.
41.
Kater AP, Owen C, Moreno C, Follows G, Munir T, Levin M-D, et al. Fixed-duration ibrutinib-venetoclax in patients with chronic lymphocytic leukemia and comorbidities. NEJM Evidence. 2022;1(7):EVIDoa2200006.
42.
Huber H, Edenhofer S, von Tresckow J, Robrecht S, Zhang C, Tausch E, et al. Obinutuzumab (GA-101), ibrutinib, and venetoclax (GIVe) frontline treatment for high-risk chronic lymphocytic leukemia. Blood. 2022 Mar 3;139(9):1318–29.
43.
Ryan C, Lampson B, Tyekucheva S, Hackett L, Ren Y, Shupe S, et al. Updated results from a multicenter, phase 2 study of acalabrutinib, venetoclax, obinutuzumab (AVO) in a population of previously untreated patients with CLL enriched for high-risk disease. Blood. 2022;140(Suppl 1):837–8.
44.
Soumerai JD, Mato AR, Dogan A, Seshan VE, Joffe E, Flaherty K, et al. Zanubrutinib, obinutuzumab, and venetoclax with minimal residual disease-driven discontinuation in previously untreated patients with chronic lymphocytic leukaemia or small lymphocytic lymphoma: a multicentre, single-arm, phase 2 trial. Lancet Haematol. 2021 Dec;8(12):e879–90.
45.
Munir T, Brown JR, O’Brien S, Barrientos JC, Barr PM, Reddy NM, et al. Final analysis from RESONATE: up to six years of follow-up on ibrutinib in patients with previously treated chronic lymphocytic leukemia or small lymphocytic lymphoma. Am J Hematol. 2019 Dec;94(12):1353–63.
46.
Byrd JC, Hillmen P, Ghia P, Kater AP, Chanan-Khan A, Furman RR, et al. Acalabrutinib versus ibrutinib in previously treated chronic lymphocytic leukemia: results of the first randomized phase III trial. J Clin Oncol. 2021 Nov 1;39(31):3441–52.
47.
Brown JR, Eichhorst B, Hillmen P, Jurczak W, Kaźmierczak M, Lamanna N, et al. Zanubrutinib or ibrutinib in relapsed or refractory chronic lymphocytic leukemia. N Engl J Med. 2023;388(4):319–32.
48.
Seymour JF, Kipps TJ, Eichhorst BF, D’Rozario J, Owen CJ, Assouline S, et al. Enduring undetectable MRD and updated outcomes in relapsed/refractory CLL after fixed-duration venetoclax-rituximab. Blood. 2022 Aug 25;140(8):839–50.
49.
Hillmen P, Rawstron AC, Brock K, Muñoz-Vicente S, Yates FJ, Bishop R, et al. Ibrutinib plus venetoclax in relapsed/refractory chronic lymphocytic leukemia: the CLARITY study. J Clin Oncol. 2019 Oct 20;37(30):2722–9.
50.
Davids M, Sharman J, Ghia P, Woyach J, Jurczak W, Siddiqi T, et al. Long-term efficacy of acalabrutinib-based regimens in patients with chronic lymphocytic leukemia and higher-risk genomic features: pooled analysis of clinical trial data. HemaSphere. 2022;6:1084–5.
51.
Rogers KA, Huang Y, Ruppert AS, Abruzzo LV, Andersen BL, Awan FT, 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 Nov 1;38(31):3626–37.
52.
Burger JA, Landau DA, Taylor-Weiner A, Bozic I, Zhang H, Sarosiek K, et al. Clonal evolution in patients with chronic lymphocytic leukaemia developing resistance to BTK inhibition. Nat Commun. 2016 May 20;7:11589.
53.
Ahn IE, Underbayev C, Albitar A, Herman SE, Tian X, Maric I, et al. Clonal evolution leading to ibrutinib resistance in chronic lymphocytic leukemia. Blood. 2017 Mar 16;129(11):1469–79.
54.
Woyach JA, Ruppert AS, Guinn D, Lehman A, Blachly JS, Lozanski A, et al. BTK(C481S)-Mediated resistance to ibrutinib in chronic lymphocytic leukemia. J Clin Oncol. 2017 May 1;35(13):1437–43.
55.
Madle S, Korte A, Bass R. Experience with mutagenicity testing of new drugs: viewpoint of a regulatory agency. Mutat Res. 1987 Aug;182(4):187–92.
56.
Zenz T, Kröber A, Scherer K, Häbe S, Bühler A, Benner A, et al. Monoallelic TP53 inactivation is associated with poor prognosis in chronic lymphocytic leukemia: results from a detailed genetic characterization with long-term follow-up. Blood. 2008 Oct 15;112(8):3322–9.
57.
Zenz T, Eichhorst B, Busch R, Denzel T, Häbe S, Winkler D, et al. TP53 mutation and survival in chronic lymphocytic leukemia. J Clin Oncol. 2010 Oct 10;28(29):4473–9.
58.
Stengel A, Kern W, Haferlach T, Meggendorfer M, Fasan A, Haferlach C. The impact of TP53 mutations and TP53 deletions on survival varies between AML, ALL, MDS and CLL: an analysis of 3307 cases. Leukemia. 2017 Mar;31(3):705–11.
59.
Brieghel C, Aarup K, Torp MH, Andersen MA, Yde CW, Tian X, et al. Clinical outcomes in patients with multi-hit TP53 chronic lymphocytic leukemia treated with ibrutinib. Clin Cancer Res. 2021 Aug 15;27(16):4531–8.
60.
Rossi D, Khiabanian H, Spina V, Ciardullo C, Bruscaggin A, Famà R, et al. Clinical impact of small TP53 mutated subclones in chronic lymphocytic leukemia. Blood. 2014 Apr 3;123(14):2139–47.
61.
Bomben R, Rossi FM, Vit F, Bittolo T, D’Agaro T, Zucchetto A, et al. TP53 mutations with low variant allele frequency predict short survival in chronic lymphocytic leukemia. Clin Cancer Res. 2021 Oct 15;27(20):5566–75.
62.
Malcikova J, Pavlova S, Kunt Vonkova B, Radova L, Plevova K, Kotaskova J, et al. Low-burden TP53 mutations in CLL: clinical impact and clonal evolution within the context of different treatment options. Blood. 2021 Dec 23;138(25):2670–85.
63.
Sun C, Nierman P, Kendall EK, Cheung J, Gulrajani M, Herman SEM, et al. Clinical and biological implications of target occupancy in CLL treated with the BTK inhibitor acalabrutinib. Blood. 2020 Jul 2;136(1):93–105.
64.
Tam CS, Trotman J, Opat S, Burger JA, Cull G, Gottlieb D, et al. Phase 1 study of the selective BTK inhibitor zanubrutinib in B-cell malignancies and safety and efficacy evaluation in CLL. Blood. 2019 Sep 12;134(11):851–9.
65.
Ahn IE, Brown JR. Targeting bruton’s tyrosine kinase in CLL. Front Immunol. 2021;12:687458.
66.
Al-Sawaf O, Zhang C, Lu T, Liao MZ, Panchal A, Robrecht S, et al. Minimal residual disease dynamics after venetoclax-obinutuzumab treatment: extended off-treatment follow-up from the randomized CLL14 study. J Clin Oncol. 2021 Dec 20;39(36):4049–60.
67.
Kater AP, Wu JQ, Kipps T, Eichhorst B, Hillmen P, D’Rozario J, 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 Dec 1;38(34):4042–54.
68.
Davids MS, Lampson BL, Tyekucheva S, Wang Z, Lowney JC, Pazienza 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 Oct;22(10):1391–402.
69.
Thompson PA, Keating MJ, Ferrajoli A, Jain N, Peterson CB, Garg N, et al. Venetoclax consolidation achieves durable off-treatment remissions in patients with high risk CLL who have been on ibrutinib more than a year. Blood. 2022;140(Supplement 1):238–40.
70.
Scarfò L, Heltai S, Albi E, Scarano E, Schiattone L, Farina L, et al. Minimal residual disease-driven treatment intensification with sequential addition of ibrutinib to venetoclax in R/R CLL. Blood. 2022 Dec 1;140(22):2348–57.
71.
Roeker LE, Feldman TA, Soumerai JD, Falco V, Panton G, Dorsey C, et al. Adding umbralisib and ublituximab (U2) to ibrutinib in patients with CLL: a phase II study of an MRD-driven approach. Clin Cancer Res. 2022 Sep 15;28(18):3958–64.
72.
Cramer P, Tausch E, von Tresckow J, Giza A, Robrecht S, Schneider C, et al. Durable remissions following combined targeted therapy in patients with CLL harboring TP53 deletions and/or mutations. Blood. 2021 Nov 11;138(19):1805–16.
73.
Döhner H, Stilgenbauer S, Benner A, Leupolt E, Kröber A, Bullinger L, et al. Genomic aberrations and survival in chronic lymphocytic leukemia. N Engl J Med. 2000 Dec 28;343(26):1910–6.
74.
Baliakas P, Jeromin S, Iskas M, Puiggros A, Plevova K, Nguyen-Khac F, et al. Cytogenetic complexity in chronic lymphocytic leukemia: definitions, associations, and clinical impact. Blood. 2019 Mar 14;133(11):1205–16.
75.
Kittai AS, Miller C, Goldstein D, Huang Y, Abruzzo LV, Beckwith K, et al. The impact of increasing karyotypic complexity and evolution on survival in patients with CLL treated with ibrutinib. Blood. 2021 Dec 9;138(23):2372–82.
76.
Rosenquist R. The more complex, the worse outcome in CLL. Blood. 2021 Dec 9;138(23):2305–7.
77.
Thompson PA, O’Brien SM, Wierda WG, Ferrajoli A, Stingo F, Smith SC, et al. Complex karyotype is a stronger predictor than del(17p) for an inferior outcome in relapsed or refractory chronic lymphocytic leukemia patients treated with ibrutinib-based regimens. Cancer. 2015;121(20):3612–21.
78.
Rigolin GM, Del Giudice I, Bardi A, Melandri A, García-Jacobo RE, Cura F, et al. Complex karyotype in unfit patients with CLL treated with ibrutinib and rituximab: the GIMEMA LLC1114 phase 2 study. Blood. 2021;138(25):2727–30.
79.
Fürstenau M, Thus Y, Robrecht S, Mellink C, van der Kevie-Kersemaekers AM, Dubois J, et al. High karyotypic complexity and translocations are adverse prognostic features in patients with chronic lymphocytic leukemia without TP53 aberrations treated with venetoclax-based time-limited combinations. Blood. 2022 11/15;140(Supplement 1):842–5.
80.
Fürstenau M, Thus YJ, Robrecht S, Mellink CHM, van der Kevie-Kersemaekers A-MF, Dubois J, et al. High karyotypic complexity is an independent prognostic factor in patients with CLL treated with venetoclax combinations. Blood. 2023;142(5):446–59.
81.
Al-Sawaf O, Lilienweiss E, Bahlo J, Robrecht S, Fink AM, Patz M, et al. High efficacy of venetoclax plus obinutuzumab in patients with complex karyotype and chronic lymphocytic leukemia. Blood. 2020 Mar 12;135(11):866–70.
82.
Anderson MA, Tam C, Lew TE, Juneja S, Juneja M, Westerman D, et al. Clinicopathological features and outcomes of progression of CLL on the BCL2 inhibitor venetoclax. Blood. 2017;129(25):3362–70.
83.
Ahn IE, Tian X, Ipe D, Cheng M, Albitar M, Tsao LC, et al. Prediction of outcome in patients with chronic lymphocytic leukemia treated with ibrutinib: development and validation of a four-factor prognostic model. J Clin Oncol. 2021 Feb 20;39(6):576–85.
84.
Chiorazzi N, Ferrarini M. Cellular origin(s) of chronic lymphocytic leukemia: cautionary notes and additional considerations and possibilities. Blood. 2011 Feb 10;117(6):1781–91.
85.
Damle RN, Wasil T, Fais F, Ghiotto F, Valetto A, Allen SL, et al. Ig V gene mutation status and CD38 expression as novel prognostic indicators in chronic lymphocytic leukemia. Blood. 1999 Sep 15;94(6):1840–7.
86.
Hamblin TJ, Davis Z, Gardiner A, Oscier DG, Stevenson FK. Unmutated Ig V(H) genes are associated with a more aggressive form of chronic lymphocytic leukemia. Blood. 1999 Sep 15;94(6):1848–54.
87.
Fischer K, Bahlo J, Fink AM, Goede V, Herling CD, Cramer P, et al. Long-term remissions after FCR chemoimmunotherapy in previously untreated patients with CLL: updated results of the CLL8 trial. Blood. 2016 Jan 14;127(2):208–15.
88.
Thompson PA, Tam CS, O’Brien SM, Wierda WG, Stingo F, Plunkett W, et al. Fludarabine, cyclophosphamide, and rituximab treatment achieves long-term disease-free survival in IGHV-mutated chronic lymphocytic leukemia. Blood. 2016 Jan 21;127(3):303–9.
89.
Tobin G, Thunberg U, Johnson A, Thörn I, Söderberg O, Hultdin M, et al. Somatically mutated Ig V(H)3-21 genes characterize a new subset of chronic lymphocytic leukemia. Blood. 2002 Mar 15;99(6):2262–4.
90.
Agathangelidis A, Darzentas N, Hadzidimitriou A, Brochet X, Murray F, Yan XJ, et al. Stereotyped B-cell receptors in one-third of chronic lymphocytic leukemia: a molecular classification with implications for targeted therapies. Blood. 2012 May 10;119(19):4467–75.
91.
Minici C, Gounari M, Übelhart R, Scarfò L, Dühren-von Minden M, Schneider D, et al. Distinct homotypic B-cell receptor interactions shape the outcome of chronic lymphocytic leukaemia. Nat Commun. 2017 Jun 9;8:15746.
92.
Nadeu F, Royo R, Clot G, Duran-Ferrer M, Navarro A, Martín S, et al. IGLV3-21R110 identifies an aggressive biological subtype of chronic lymphocytic leukemia with intermediate epigenetics. Blood. 2021 May 27;137(21):2935–46.
93.
Burger JA, Tedeschi A, Barr PM, Robak T, Owen C, Ghia P, et al. Ibrutinib as initial therapy for patients with chronic lymphocytic leukemia. N Engl J Med. 2015 Dec 17;373(25):2425–37.
94.
Munir T, Pitchford A, Bloor A, Pettitt A, Patten P, Forconi F, et al. Combination of ibrutinib plus venetoclax with MRD-driven duration of treatment results in a higher rate of MRD negativity in IGHV unmutated than mutated CLL: updated interim analysis of FLAIR study. Blood. 2022 11/15;140(Supplement 1):231–3.
95.
Niemann C, Munir T, Moreno C, Owen C, Follows G, Benjamini O, et al. Residual disease kinetics among patients with high-risk factors treated with first-line fixed-duration ibrutinib plus venetoclax (Ibr + Ven) versus chlorambucil plus obinutuzumab (Clb+O): the glow study. Blood. 2022 11/15;140(Supplement 1):228–30.
96.
Fürstenau M, De Silva N, Eichhorst B, Hallek M. Minimal residual disease assessment in CLL: ready for use in clinical routine?Hemasphere. 2019 Oct;3(5):e287.
97.
Böttcher S, Ritgen M, Fischer K, Stilgenbauer S, Busch RM, Fingerle-Rowson G, 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 Mar 20;30(9):980–8.
98.
Strati P, Keating MJ, O’Brien SM, Burger J, Ferrajoli A, Jain N, et al. Eradication of bone marrow minimal residual disease may prompt early treatment discontinuation in CLL. Blood. 2014 Jun 12;123(24):3727–32.
99.
Moreno C, Greil R, Demirkan F, Tedeschi A, Anz B, Larratt L, et al. First-line treatment of chronic lymphocytic leukemia with ibrutinib plus obinutuzumab versus chlorambucil plus obinutuzumab: final analysis of the randomized, phase III iLLUMINATE trial. Haematologica. 2022 Sep 1;107(9):2108–20.
100.
Davids MS, Brander DM, Kim HT, Tyekucheva S, Bsat J, Savell A, 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 Aug;6(8):e419–28.
101.
Fraser G, Cramer P, Demirkan F, Silva RS, Grosicki S, Pristupa A, 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 Apr;33(4):969–80.
102.
Jain N, Thompson P, Burger J, Ferrajoli A, Takahashi K, Estrov Z, et al. Ibrutinib, fludarabine, cyclophosphamide, and obinutuzumab (iFCG) regimen for chronic lymphocytic leukemia (CLL) with mutated IGHV and without TP53 aberrations. Leukemia. 2021 Dec;35(12):3421–9.
103.
Sharman JP, Egyed M, Jurczak W, Skarbnik A, Pagel JM, Flinn IW, et al. Efficacy and safety in a 4-year follow-up of the ELEVATE-TN study comparing acalabrutinib with or without obinutuzumab versus obinutuzumab plus chlorambucil in treatment-naïve chronic lymphocytic leukemia. Leukemia. 2022 Apr;36(4):1171–5.
104.
Soumerai JD, Ni A, Darif M, Londhe A, Xing G, Mun Y, et al. Prognostic risk score for patients with relapsed or refractory chronic lymphocytic leukaemia treated with targeted therapies or chemoimmunotherapy: a retrospective, pooled cohort study with external validations. Lancet Haematol. 2019 Jul;6(7):e366–74.
105.
Liu Y-J, Arpin C. Germinal center development. Immunol Rev. 1997;156(1):111–26.
106.
Rose DM, Han J, Ginsberg MH. Alpha4 integrins and the immune response. Immunol Rev. 2002 Aug;186:118–24.
107.
Burger JA. Targeting the microenvironment in chronic lymphocytic leukemia is changing the therapeutic landscape. Curr Opin Oncol. 2012 Nov;24(6):643–9.
108.
Tissino E, Benedetti D, Herman SEM, Ten Hacken E, Ahn IE, Chaffee KG, et al. Functional and clinical relevance of VLA-4 (CD49d/CD29) in ibrutinib-treated chronic lymphocytic leukemia. J Exp Med. 2018 Feb 5;215(2):681–97.
109.
Bulian P, Shanafelt TD, Fegan C, Zucchetto A, Cro L, Nückel H, et al. CD49d is the strongest flow cytometry-based predictor of overall survival in chronic lymphocytic leukemia. J Clin Oncol. 2014 Mar 20;32(9):897–904.
110.
Taylor AM, Metcalfe JA, Thick J, Mak YF. Leukemia and lymphoma in ataxia telangiectasia. Blood. 1996 Jan 15;87(2):423–38.
111.
Neilson JR, Auer R, White D, Bienz N, Waters JJ, Whittaker JA, et al. Deletions at 11q identify a subset of patients with typical CLL who show consistent disease progression and reduced survival. Leukemia. 1997 Nov;11(11):1929–32.
112.
Kröber A, Seiler T, Benner A, Bullinger L, Brückle E, Lichter P, et al. V(H) mutation status, CD38 expression level, genomic aberrations, and survival in chronic lymphocytic leukemia. Blood. 2002 Aug 15;100(4):1410–6.
113.
Rossi D, Bruscaggin A, Spina V, Rasi S, Khiabanian H, Messina M, et al. Mutations of the SF3B1 splicing factor in chronic lymphocytic leukemia: association with progression and fludarabine-refractoriness. Blood. 2011 Dec 22;118(26):6904–8.
114.
Kipps TJ, Fraser G, Coutre SE, Brown JR, Barrientos JC, Barr PM, et al. Long-term studies assessing outcomes of ibrutinib therapy in patients with del(11q) chronic lymphocytic leukemia. Clin Lymphoma Myeloma Leuk. 2019 Nov;19(11):715–22.e6.
115.
Wierda WG, O’Brien S, Wang X, Faderl S, Ferrajoli A, Do KA, et al. Prognostic nomogram and index for overall survival in previously untreated patients with chronic lymphocytic leukemia. Blood. 2007 Jun 1;109(11):4679–85.
116.
Jaksić B, Vitale B, Hauptmann E, Planinc-Peraica A, Ostojic S, Kusec R. The roles of age and sex in the prognosis of chronic leukaemias. A study of 373 cases. Br J Cancer. 1991 Aug;64(2):345–8.
117.
Shanafelt TD, Rabe KG, Kay NE, Zent CS, Jelinek DF, Reinalda MS, et al. Age at diagnosis and the utility of prognostic testing in patients with chronic lymphocytic leukemia. Cancer. 2010 Oct 15;116(20):4777–87.
118.
Gordon MJ, Kaempf A, Sitlinger A, Shouse G, Mei M, Brander DM, et al. The chronic lymphocytic leukemia comorbidity index (CLL-CI): a three-factor comorbidity model. Clin Cancer Res. 2021 Sep 1;27(17):4814–24.
119.
Rotbain EC, Gordon MJ, Vainer N, Frederiksen H, Hjalgrim H, Danilov AV, et al. The CLL comorbidity index in a population-based cohort: a tool for clinical care and research. Blood Adv. 2022 Apr 26;6(8):2701–6.
120.
Eichhorst B, Goede V, Hallek M. Treatment of elderly patients with chronic lymphocytic leukemia. Leuk Lymphoma. 2009 Feb;50(2):171–8.
121.
Ahn IE. Cardiovascular adverse events of ibrutinib. Blood. 2019 Nov 28;134(22):1881–2.
122.
Dickerson T, Wiczer T, Waller A, Philippon J, Porter K, Haddad D, et al. Hypertension and incident cardiovascular events following ibrutinib initiation. Blood. 2019 Nov 28;134(22):1919–28.
123.
Bhat SA, Gambril J, Azali L, Chen ST, Rosen L, Palettas M, et al. Ventricular arrhythmias and sudden death events following acalabrutinib initiation. Blood. 2022 Nov 17;140(20):2142–5.
124.
Lipsky AH, Farooqui MZ, Tian X, Martyr S, Cullinane AM, Nghiem K, et al. Incidence and risk factors of bleeding-related adverse events in patients with chronic lymphocytic leukemia treated with ibrutinib. Haematologica. 2015 Dec;100(12):1571–8.