Modern treatment approaches such as chemoimmunotherapy (e.g., fludarabine/cyclophosphamide/rituximab or FCR) are highly effective in the majority of chronic lymphocytic leukemia (CLL) patients. However, there remains a small but challenging subgroup of patients who show ultra high-risk genetics (17p deletion, TP53 mutation) and/or poor response to chemoimmunotherapy. The median life expectancy of these patients is below 2 to 3 years with standard regimens. Accordingly, CLL with the 17p deletion (and likely also with sole TP53 mutation) should be treated with alternative strategies. While p53 defects appear to play a central role in our understanding of this ultra high-risk group, at least half of the cases will not be predictable based on existing prognostic models. Current treatment approaches for patients with p53 defects or poor response to chemoimmunotherapy should rely on agents acting independently of p53, such as alemtuzumab, lenalidomide, flavopiridol, and a growing number of novel compounds (or combinations thereof) currently available in clinical trials. Poor survival times of patients with ultra high-risk CLL suggest that eligible patients should be offered consolidation with reduced-intensity allogeneic stem-cell transplantation or experimental approaches in clinical trials.

In contrast to most other types of leukemia, chronic lymphocytic leukemia (CLL) is not treated upon diagnosis, but upon progression to symptomatic disease.1  This approach is based on the results of randomized trials failing to show benefits of unstratified early treatment with chemotherapy. However, over the last decades of CLL treatment, we have seen the transition from single-agent, alkylator-based therapies to nucleoside analogs, combination chemotherapy, and, most recently, chemoimmunotherapy (e.g., combining monoclonal antibodies such as rituximab with chemotherapy such as fludarabine + cyclophosphamide, which is called FCR).2  As a result, complete response rates have improved from 3% to 7% with chlorambucil to a maximum of 70% with FCR.3–7  In the randomized German CLL Study Group (GCLLSG) CLL8 trial comparing FC and FCR, the complete response rate in the FCR arm was 44%.5  About 90% to 95% of patients receiving FCR as a first-line treatment are expected to respond. Historical comparisons suggest that for the first time, overall survival (OS) has been improved with the application of chemoimmunotherapy.8,9  In addition, recent results suggest that modern therapies are changing the natural course of CLL: multivariate analysis including genetics (FISH, TP53 mutation, IGHV) within prospective trials (GCLLSG CLL4: F vs. FC trial), and, most recently, results of the CLL8 (FC vs. FCR) trial suggest that the initial treatment choices influence OS. However, in all available analyses, there is a strong association between genetic disease profile and outcome.10  Therefore, the development of prognostic and predictive models leading to stratified treatment strategies for specific disease subgroups are of importance. While most patients with CLL will either not need treatment or will respond well to treatment, there remains a group of patients with refractory CLL, as defined by the International Workshop on Chronic Lymphocytic Leukemia (IWCLL) and the National Cancer Institute (NCI) working groups,11 ) or suboptimal response (early relapse after exposure to FCR or regimens of similar potency). Identifying this group of patients early, ideally before treatment, by clinical or biological parameters and designing adequate treatment options remain key issues in the treatment of CLL.12  This will benefit not only the ultra high-risk group but other patients as well, because targeted approaches can also be curtailed to define low-risk groups. For this review, we have defined ultra high-risk CLL as patient subgroups with median survival below 24 to 36 months at the treatment time point. Based on this definition, patients with the 17p deletion, the TP53 mutation, and F-refractory CLL belong to this group, as well as patients with suboptimal response to intense treatment (e.g., FCR) (Figure 1).

Figure 1.

Definition of ultra high-risk CLL and incidence of the specific subgroups. The definitions for the purpose of this article are given with regard to biological (i.e., 17p deletion, TP53 mutation) and clinical (i.e., response to treatment) characteristics.

Figure 1.

Definition of ultra high-risk CLL and incidence of the specific subgroups. The definitions for the purpose of this article are given with regard to biological (i.e., 17p deletion, TP53 mutation) and clinical (i.e., response to treatment) characteristics.

Close modal

It is important to stress that other prognostic factors associated with inferior outcome in CLL, such as the 11q deletion, unmutated IGHV, ZAP70 expression, or ß2-microglobulin levels, will not be discussed here. While these are clearly associated with poor prognosis, they cannot be included in the ultra high-risk category of patients treated with chemoimmunotherapy according to the definition given above.5,13 

Molecular Mechanisms Underlying Refractory CLL

p53 plays a central role in our current understanding of why CLL patients fail to respond to chemo(immuno)therapy and have short survival times. The single best-characterized molecular lesions found in refractory CLL are the deletion 17p13 (TP53 locus) and TP53 mutations.6,14–17 

Poor outcome of patients with the 17p deletion has been observed for over 15 years, and the initial single-center experience documenting the failure to induce remissions with purine analog-based chemotherapy in this group of patients18  has been confirmed in all prospective trials since.3,14,19  Indeed, this group of patients (5%–8% of patients receiving first-line treatment)13,14,19,20  are the first group to be treated in separate (genotype-specific) treatment trials in CLL. This is based on the observation that median OS will be in the range of 2 to 3 years from the time of front-line treatment. While there is a small subgroup of patients with the 17p deletion (and mostly mutated IGHV) without treatment indications who may exhibit stable disease for years,21,22  the outcome of most patients with the 17p deletion and need for treatment is very poor.13,14,19,20  Patients with the 17p deletion will very rarely achieve complete response after chemoimmunotherapy, as demonstrated in the CLL8 trial, in which the group without the 17p deletion had a complete response rate of 21.8% (FC) or 44.1% (FCR) compared with 2.3% among patients with the 17p deletion (both arms).5 

Over 80% of CLL patients with the 17p deletion also carry a TP53 mutation on the remaining allele.23–25  There have been a number of retrospective studies suggesting that, in addition to the 17p deletion, the TP53 mutation even in the absence of the 17p deletion predicts poor outcome.24–27  In a recent analysis of the GCLLSG CLL4 trial (F vs. FC), we found TP53 mutations in 8.5% of patients (28 of 328; 4.5% in the absence of the 17p deletion).23  No complete response was observed in patients with the TP53 mutation. Median progression-free survival (PFS) (23.3 vs. 62.2 months) and OS (29.2 vs. 84.6 months) were significantly decreased in the group with the TP53 mutation (both p < 0.001). PFS and OS for patients with the 17p deletion and patients with the TP53 mutation in the absence of the 17p deletion were similar (Figure 2). Multivariate analysis identified the TP53 mutation as the strongest prognostic marker regarding OS (hazard ratio [HR] 7.2, p < 0.001). Other independent predictors of OS were unmutated IGHV status (HR 1.9), 11q deletion (HR 1.9), 17p deletion (HR 2.3), and FC treatment (HR 0.6).

Figure 2.

CLL with TP53 mutation without the 17p deletion shows similar outcome as CLL with the 17p deletion after first-line F-based therapy (CLL4 trial of the GCLLSG). OS of the group with the 17p deletion (n = 16) is shown in red; OS of the sole TP53 mutation without the 17p deletion (n = 14) is shown in black; and the OS of remaining patients (n = 277) is shown in green. Median OS was significantly shorter for patients with the 17p deletion (19.2 months) and sole TP53 mutation (30.2 months) compared with patients without either abnormality (median not reached; p < 0.001). (Data from 23 .)

Figure 2.

CLL with TP53 mutation without the 17p deletion shows similar outcome as CLL with the 17p deletion after first-line F-based therapy (CLL4 trial of the GCLLSG). OS of the group with the 17p deletion (n = 16) is shown in red; OS of the sole TP53 mutation without the 17p deletion (n = 14) is shown in black; and the OS of remaining patients (n = 277) is shown in green. Median OS was significantly shorter for patients with the 17p deletion (19.2 months) and sole TP53 mutation (30.2 months) compared with patients without either abnormality (median not reached; p < 0.001). (Data from 23 .)

Close modal

Early results from the CLL8 trial suggest that patients with the TP53 mutation only, the 17p deletion without the TP53 mutation, and both the mutation and the deletion have poor response to FC and FCR.28  In contrast, data from the ECOG 2997 trial and the UK LRF CLL4 trial did not show such profound effects of the TP53 mutation in the absence of the 17p deletion, but technical approaches and incidence of mutations and the choice of treatment (UK LRF4) were different.14,29  In general, it appears that the impact of biological prognostic markers such as the TP53 mutation and the 17p deletion is the most pronounced when more potent treatments (i.e., FC or FCR) are applied, likely due to the fact that the outcome of most subgroups is markedly improved, while the outcome of the ultra high-risk patients remains essentially unchanged. Further evidence for the resistance of clones with the 17p deletion and the TP53 mutation comes from the observation that these aberrations are the most common abnormalities acquired during the course of the disease (clonal evolution), and, importantly, expand under the selection pressure of chemo(immuno)therapy.24,30,31  With advanced treatment lines, the incidence of TP53 aberrations increases from approximately 10% (first-line), to approximately 20% at relapse and as much as 40% to 50% in F-refractory CLL.10,17 

Because CLL with the TP53 mutation carries a very poor prognosis regardless of the presence of the 17p deletion when treated with F-based chemotherapy, TP53 mutation analysis should be incorporated into the evaluation of CLL patients before treatment initiation. CLL patients with the TP53 mutation and indication for therapy should be considered for alternative treatment approaches up front because they are at ultra high risk. These findings are of particular importance because the group of patients who should be considered for alternative approaches (genotype-specific) will increase from 5% to 8% up to 8% to 12% for front-line therapy when considering the TP53 mutation in addition to the 17p deletion.23,28 

Even in spite of the strong association between poor clinical outcome and the TP53 mutation, p53 abnormalities will only explain a fraction of refractory CLL cases. The proportion of F-refractory CLL cases carrying the TP53 loss or mutation has been estimated to add up to 40 to 50.16,17  Current data suggest that 29% to 52% of F-refractory CLL cases (treated with F or FC/FCR) in first-line-treatment settings may be explained by the TP53 mutation and/or the 17p deletion.23,28 

These findings raise questions regarding the basis of refractoriness in the remaining cases. While there are numerous studies showing defects in the DNA-damage machinery in CLL, this issue has not been adequately addressed in larger studies. There is some evidence that additional cases are associated with p53 pathway defects such as miR-34a dysregulation,32  but the mechanisms of refractory CLL remain an important research question that is best addressed by diverse biological analyses of carefully annotated samples from clinical trials.

Clinical Definition of Ultra High-Risk CLL Based on Response

While it is important to identify new prognostic and predictive factors, there is a clear picture emerging that, even in the absence of known ultra high-risk markers, poor response to intensive therapy (e.g., FCR) will be associated with short OS and can be considered ultra high risk. Results from the M.D. Anderson Cancer Center (MDACC)8,33  (Keating et al., personal communication) and from the CLL8 trial (FC vs. FCR) suggest that OS is very poor for patients who need early (within 2–3 years) re-treatment after first-line therapy.

In the pivotal trial cohort from MDACC establishing FCR as the treatment with the highest efficacy in CLL,4,8  patients with a PFS below 36 months had a significantly shorter OS (<24 months from subsequent treatment) compared with patients with a PFS > 36 months (p < 0.01) (Keating et al., personal communication). While this was a single-institution trial, very similar results are becoming available from randomized multi-center trials. In an analysis from the GCLLSG CLL8 trial (FC and FCR arm), patients with a PFS below 12 months (n = 47), 12 to 24 months (n = 45), or above 24 months showed significant differences in OS from the time point of second-line treatment (median 13.1, 20.3, and 44.6 months, respectively; p < 0.01) (Figure 3). These data suggest that patients with PFS below 24 months should be considered ultra high risk at the time of recurring treatment indication. It is important to note that similar outcome can be expected in patients treated with comparable regimens incorporating other chemotherapy agents such as pentostatin, mitoxantrone, and bendamustine (e.g. PCR, FCM-R, BR).

Figure 3.

Patients relapsing early after FC or FCR have a poor outcome. OS from the time point of second-line treatment for patients initially treated in the CLL8 trial (FC vs. FCR) according to remission duration after first-line treatment. Patients with short remissions (<24 months) have very poor outcomes, suggesting that these patients should be considered for investigational treatment and allo-SCT.

Figure 3.

Patients relapsing early after FC or FCR have a poor outcome. OS from the time point of second-line treatment for patients initially treated in the CLL8 trial (FC vs. FCR) according to remission duration after first-line treatment. Patients with short remissions (<24 months) have very poor outcomes, suggesting that these patients should be considered for investigational treatment and allo-SCT.

Close modal

While these observations will only be relevant to a small subgroup of patients (about 10% of patients treated in first-line; e.g., 92 of 814 [11.3%] in the CLL8 trial), it is an important group of patients. In the design of trials, this group should be addressed separately from late relapses or relapses after non-intense pretreatment. In many ways, the classical definition of F-refractory CLL (no remission or remission under 6 months in duration1 ) and corresponding risk evaluation is currently undergoing reassessment in the era of chemoimmunotherapy.

Targeted Treatment/Genotype-Specific Treatment in CLL

Ideally, the treatment approach should target the molecular lesion critical in ultra high-risk CLL. This can be envisioned in cases of CLL with mutant TP53. Remarkable progress has been made in the development of p53-binding molecules that can rescue the function of certain p53 mutants. Agents have been identified that exploit the p53 pathway by either seeking targets and compounds that show synthetic lethality with TP53 mutations or by looking for non-genotoxic activators of the p53 response.34  While there is some evidence that cells with mutant TP53 may exhibit specific vulnerabilities, clinical translation of this is so far inconclusive.34–36  The approach currently taken for CLL with loss or mutation of TP53 and F-refractory CLL relies mostly on the use of agents acting independently of the DNA damage pathway.37  While the success of this approach has been documented for antibodies (e.g., alemtuzumab), there is still considerable room for improvement. Most current treatment recommendations or approvals from regulatory authorities are based on non-randomized trials.38,39  There is little evidence that patients with the 17p deletion/TP53 mutation or FCR-refractory CLL will benefit from alternative chemotherapy regimens, because clinical trials show similar outcomes for these patients after treatment such as BR or R-FCM.40,41 

As shown in Table 1, most regimes used in the setting of CLL with the 17p deletion or F-refractoriness can be expected to induce responses in 30% to 50% of patients with relatively short PFS. Based on these results and the data presented above, these patient groups are prime candidates for novel clinical trials (Figure 4). Ideally, these should be randomized or multi-arm studies using novel agents for which there is at least some evidence for specific activity in ultra high-risk CLL (Table 1).

Table 1.

ORR and PFS for patients with the 17p deletion or F-refractory CLL after different treatment approaches

ORR and PFS for patients with the 17p deletion or F-refractory CLL after different treatment approaches
ORR and PFS for patients with the 17p deletion or F-refractory CLL after different treatment approaches

#Data shown are only for patients with F-refractory CLL.

Figure 4.

Suggested definitions and corresponding treatment algorithms for patients with ultra high-risk CLL. Three groups are defined: 1) patients with the 17p deletion or the TP53 mutation and treatment indication, 2) patients with F-refractory CLL, and 3) patients with short PFS after intense therapy (e.g. FCR). Treatment results with current “standard” chemo(immuno)therapy are poor, and therefore these patients are prime candidates for clinical trials evaluating novel agents (that would ideally be comparative). The choice of agent/trial should be based on evidence for efficacy available in the specific situation (i.e., the TP53 mutation). In addition, these patients are prime candidates for consolidation with reduced intensity allo-SCT (fit patients) or experimental approaches in clinical trials. These suggestions are rarely based on comparative trial data because these are scarce.

Figure 4.

Suggested definitions and corresponding treatment algorithms for patients with ultra high-risk CLL. Three groups are defined: 1) patients with the 17p deletion or the TP53 mutation and treatment indication, 2) patients with F-refractory CLL, and 3) patients with short PFS after intense therapy (e.g. FCR). Treatment results with current “standard” chemo(immuno)therapy are poor, and therefore these patients are prime candidates for clinical trials evaluating novel agents (that would ideally be comparative). The choice of agent/trial should be based on evidence for efficacy available in the specific situation (i.e., the TP53 mutation). In addition, these patients are prime candidates for consolidation with reduced intensity allo-SCT (fit patients) or experimental approaches in clinical trials. These suggestions are rarely based on comparative trial data because these are scarce.

Close modal

There are numerous targeted strategies aiming at key pathways (e.g. PI3-kinase, NFkB, SYK, AKT) and a number of antibody-based approaches targeting alternative epitopes (e.g., CD19, CD20, CD23, CD37) currently available in clinical trials. A detailed discussion of these is beyond the scope of this article. Figure 5 gives an overview of a selection of substances in clinical development in CLL.

Figure 5.

Selection of targets and novel agents in early clinical trials of CLL. There are multiple “biological” therapeutic approaches in current clinical development in CLL. Signal transduction inhibition will often target the B-cell receptor cascade, which has been shown to be critical to CLL cell survival. A number of surface molecules of CLL cells can be targeted by antibodies. Different members of the apoptosis machinery are targets currently explored in clinical trials. Microenvironmental stimulation and T-cell interaction are of critical importance for CLL cell survival, and are being increasingly utilized for treatment strategies. There is ongoing interest in using mutant p53 as a “druggable” target, and a number of agents have been identified that may preferentially target mutant p53.34  Improving efficacy in CLL with p53 pathway defects is likely to offer the greatest overall benefit.

Figure 5.

Selection of targets and novel agents in early clinical trials of CLL. There are multiple “biological” therapeutic approaches in current clinical development in CLL. Signal transduction inhibition will often target the B-cell receptor cascade, which has been shown to be critical to CLL cell survival. A number of surface molecules of CLL cells can be targeted by antibodies. Different members of the apoptosis machinery are targets currently explored in clinical trials. Microenvironmental stimulation and T-cell interaction are of critical importance for CLL cell survival, and are being increasingly utilized for treatment strategies. There is ongoing interest in using mutant p53 as a “druggable” target, and a number of agents have been identified that may preferentially target mutant p53.34  Improving efficacy in CLL with p53 pathway defects is likely to offer the greatest overall benefit.

Close modal

Treatment of Patients with F-Refractory CLL

Patients with F-refractory CLL, today mostly referring to F-based chemo(immuno)therapy combinations, have a very poor prognosis.42  Response to various chemotherapeutic approaches is low in this setting, and alternative approaches (acting independently of the DNA damage pathway) are preferred (Figures 4 and 5). The anti-CD52 antibody alemtuzumab is well studied in patients with refractory CLL, and is documented to induce remission independent of the genetic make-up of the CLL cells.43,44  Responses can be expected in about one-third of patients with F-refractory CLL and in 60% to 80% of treatment-naive patients; however, remission duration is short if no consolidation strategies are applied.

There is relatively little information on the use of rituximab in F-refractory CLL, but it is used in combination with a number of agents this setting (e.g., steroids). Recently, the novel CD20 antibody ofatumumab has been assessed in a phase II study of patients with F- or F + alemtuzumab-refractory CLL. Results showed an encouraging 50% overall response rate (ORR; assessed during ongoing treatment), but a relatively short PFS of 5.9 months.38  Activity in CLL with the 17p deletion has not been clearly demonstrated.

Flavopiridol is a broad, cyclin-dependent kinase inhibitor that induces apoptosis in leukemic cell lines and CLL cells, and its activity is p53 independent. In the pivotal study by Byrd et al., 45% of patients with high-risk CLL responded, and their PFS was 12 months.45  Currently, flavopiridol is being evaluated in an international multicenter phase II study of F-refractory CLL to further explore its use in this setting.

Lenalidomide has been used in advanced CLL. Much of the agent's appeal is based on a mechanism of action that is, even if incompletely understood, very different from chemotherapy or other agents.46  Lenalidomide has been studied in a limited number of patients with the 17p deletion or F-refractory disease, and the results are summarized in Table 1. Current approaches with this agent explore combinations and maintenance/consolidation strategies.

Treatment of Patients with Suboptimal Response to Intensive Chemoimmunotherapy

Similarly to OS, which correlates well with remission duration after FCR, the ORR after second-line therapy will depend on the length of initial remission. In an analysis of the response to subsequent treatment of patients in the initial FCR trial,4,8  the ORR to intense second-line treatment was 44% (PFS after FCR < 36 months) compared with 70% (>36 months) (Keating et al., personal communication). The clinical consequences of this observation should influence the choice of relapse treatment and consolidation approaches (e.g., allogeneic stem-cell transplantation [allo-SCT]). Based on results from MDACC, a potential approach to these patients (PFS < 24–36 months after initial FCR) would be to consider re-treatment with FCR or similar regimens in patients who responded to first-line treatment with a complete response or nodular partial response, because the ORR is 68% to 78% in these patients. On the other hand, patients who only had a partial response after first-line FCR treatment should be treated with investigational agents, because responses will be in the range of only 30% (M. Keating et al., personal communication) (Figure 4). The results of CLL8 confirm that patients with short response durations (<24 months) after initial treatment with FC or FCR should be considered candidates for trials investigating alternative approaches (Figures 3, 4, and 5).

Currently, evidence is accumulating from prospective trials that a refined guidance of treatment will be possible by assessing minimal residual disease (MRD). A number of studies have shown the association of improved outcome with MRD negativity.47,48  Correspondingly, high MRD levels correlate with poor outcome after chemo(immuno)therapy, and it is foreseeable that MRD assessment could allow further risk stratification early during or after treatment to identify high-risk or ultra high-risk patients.49 

Allo-SCT

The recent past has seen a surge in transplantations for patients with CLL. This has been supported by the use of reduced-intensity conditioning regimens, improved (genetic) risk stratification, and the availability of more data on outcome after allo-SCT. A recent proposal by international experts has defined a consensus on when to consider allo-SCT in CLL.50  Although “poor-risk CLL” had not been defined unequivocally at that time, allo-SCT was proposed for younger patients who do not respond to or relapse early (< 12 months) after therapy with purine analog, those who relapse within 24 months after purine analog-based combination therapy or autologous transplantation, and those with TP53 abnormalities (deletions or mutations) requiring treatment. Indeed, based on the recent data described above, this recommendation can be fully endorsed.

Clinical trial data suggest that the 4-year OS of high-risk CLL patients receiving a reduced-intensity allo-SCT is between 48% and 70%.51  In the setting of allo-SCT, the ultra high-risk features 17p deletion or F-refractoriness appear not to affect efficacy per se. In contrast, disease status at the time of SCT (i.e., lack of remission or bulky lymphadenopathy) strongly predicts failure. This again underlines the need for alternative approaches for remission induction in ultra high-risk CLL patients to make reduced-intensity allo-SCT a feasible option. Prospective trials are warranted to define the optimal transplant strategies best suited for different clinical scenarios. In addition, there is an urgent need to explore non-transplant consolidation options, because the majority of ultra high-risk CLL patients will not be transplant candidates due to their age and comorbidities.

We currently consider patients with the 17p deletion or TP53 mutation who need treatment the only established ultra high-risk CLL group based on biological markers. However, there is growing evidence that the group of patients with F-refractory disease or early relapse (<24–36 months) after FCR or similar treatment should also be considered ultra high risk. Ultra high-risk patients should be considered for treatment trials with novel compounds and combinations, and they are prime candidates for allo-SCT as consolidation. Further investigation of other biological causes in addition to p53 defects is of prime importance not only to enhance our understanding of ultra high-risk CLL, but also to help in its management.

We are indebted to Michael Keating for sharing unpublished results and helpful discussions vital to the manuscript. We thank Asher Chanan-Khan for sharing unpublished data and Peter Dreger, Andreas Viardot, Michael Hallek, Alessandra Ferrajoli, John Byrd, and Michael Keating for critical reading of the manuscript. We wish to acknowledge the important contributions of the numerous investigators whose work could not be cited due to space restrictions. We acknowledge the GCLLSG (chairman Michael Hallek) and the European Research Initiative on CLL (ERIC, chairman E. Montserrat) for support and discussions. We thank Hartmut Döhner for longstanding support and mentoring.

Conflict-of-interest disclosure: SS has consulted for and received research funding and honoraria from Roche, Mundipharma, GSK, Genzyme, Boehringer-Ingelheim, Bayer, Amgen, and Sanofi Aventis. TZ has been a member of advisory boards and has received honoraria from Boehringer-Ingelheim, Celgene, GSK, Trubion, and Bayer-Schering Pharma. Off-label drug use: Off-label use of diagnostic tests and therapeutic agents.

Funded in part by the DFG (DFG STI 296/2–1), German José Carreras Leukemia-Foundation (R06/28v, R08/26f), Else Kröner-Fresenius-Stiftung (P20/07//A11/07), Deutsche Krebshilfe (108355, 106142), and the Global CLL Research Foundation.

Dr. Stephan Stilgenbauer, Department of Internal Medicine III, University of Ulm, Albert-Einstein-Allee 23, 89081 Ulm, Germany; Phone: 49–731-500–45521; Fax: 49–731-500–45525; e-mail: stephan.stilgenbauer@uniklinik-ulm.de

1
Hallek
M
Cheson
BD
Catovsky
D
et al
Guidelines for the diagnosis and treatment of chronic lymphocytic leukemia: a report from the International Workshop on Chronic Lymphocytic Leukemia updating the National Cancer Institute-Working Group 1996 guidelines
Blood
2008
111
5446
5456
2
Gribben
JG
How I treat CLL up front
Blood
2010
115
187
197
3
Catovsky
D
Richards
S
Matutes
E
et al
Assessment of fludarabine plus cyclophosphamide for patients with chronic lymphocytic leukaemia (the LRF CLL4 Trial): a randomised controlled trial
Lancet
2007
370
230
239
4
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
23
4079
4088
5
Hallek
M
Fingerle-Rowson
G
Fink
A-M
et al
First-Line Treatment with Fludarabine (F), Cyclophosphamide (C), and Rituximab (R) (FCR) Improves Overall Survival (OS) in Previously Untreated Patients (pts) with Advanced Chronic Lymphocytic Leukemia (CLL): Results of a Randomized Phase III Trial On Behalf of An International Group of Investigators and the German CLL Study Group [Abstract]
Blood
2009
114
22
535
6
Eichhorst
BF
Busch
R
Hopfinger
G
et al
Fludarabine plus cyclophosphamide versus fludarabine alone in first-line therapy of younger patients with chronic lymphocytic leukemia
Blood
2006
107
885
891
7
Flinn
IW
Neuberg
DS
Grever
MR
et al
Phase III trial of fludarabine plus cyclophosphamide compared with fludarabine for patients with previously untreated chronic lymphocytic leukemia: US Intergroup Trial E2997
J Clin Oncol
2007
25
793
798
8
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
112
975
980
9
Abrisqueta
P
Pereira
A
Rozman
C
et al
Improving survival in patients with chronic lymphocytic leukemia (1980–2008): the Hospital Clinic of Barcelona experience
Blood
2009
114
10
2044
2050
10
Zenz
T
Mertens
D
Küppers
R
Döhner
H
Stilgenbauer
S
From pathogenesis to treatment of chronic lymphocytic leukaemia
Nat Rev Cancer
2010
1
10
1
37
50
11
Cheson
BD
Bennett
JM
Grever
M
et al
National Cancer Institute-sponsored Working Group guidelines for chronic lymphocytic leukemia: revised guidelines for diagnosis and treatment
Blood
1996
87
4990
4997
12
Montserrat
E
Moreno
C
Esteve
J
Urbano-Ispizua
A
Gine
E
Bosch
F
How I treat refractory CLL
Blood
2006
107
1276
1283
13
Oscier
D
Wade
R
Davis
Z
et al
Prognostic factors identify 3 risk groups in the LRF CLL4 trial, independent of treatment allocation
Haematologica
2010
10
95
10
1705
1712
14
Grever
MR
Lucas
DM
Dewald
GW
et al
Comprehensive assessment of genetic and molecular features predicting outcome in patients with chronic lymphocytic leukemia: results from the US Intergroup Phase III Trial E2997
J Clin Oncol
2007
25
799
804
15
Byrd
JC
Gribben
JG
Peterson
BL
et al
Select high-risk genetic features predict earlier progression following chemoimmunotherapy with fludarabine and rituximab in chronic lymphocytic leukemia: justification for risk-adapted therapy
J Clin Oncol
2006
24
437
443
16
Lozanski
G
Heerema
NA
Flinn
IW
et al
Alemtuzumab is an effective therapy for chronic lymphocytic leukemia with p53 mutations and deletions
Blood
2004
103
3278
3281
17
Zenz
T
Häbe
S
Denzel
T
et al
Detailed analysis of p53 pathway defects in fludarabine-refractory chronic lymphocytic leukemia (CLL): dissecting the contribution of 17p deletion, TP53 mutation, p53-p21 dysfunction, and miR34a in a prospective clinical trial
Blood
2009
114
2589
2597
18
Döhner
H
Fischer
K
Bentz
M
et al
p53 gene deletion predicts for poor survival and non-response to therapy with purine analogs in chronic B-cell leukemias
Blood
1995
85
1580
1589
19
Stilgenbauer
S
Eichhorst
BF
Busch
R
et al
Biologic and Clinical Markers for Outcome after Fludarabine (F) or F Plus Cyclophosphamide (FC) - Comprehensive Analysis of the CLL4 Trial of the GCLLSG [Abstract]
Blood
2008
112
11
2089
20
Stilgenbauer
S
Zenz
T
Winkler
D
et al
Genomic Aberrations, VH Mutation Status and Outcome after Fludarabine and Cyclophosphamide (FC) or FC Plus Rituximab (FCR) in the CLL8 Trial [Abstract]
Blood
2008
112
11
781
21
Best
OG
Gardiner
AC
Davis
ZA
et al
A subset of Binet stage A CLL patients with TP53 abnormalities and mutated IGHV genes have stable disease
Leukemia
2009
23
212
214
22
Tam
CS
Shanafelt
TD
Wierda
WG
et al
De novo deletion 17p13.1 chronic lymphocytic leukemia shows significant clinical heterogeneity: the M. D. Anderson and Mayo Clinic experience
Blood
2009
114
957
964
23
Zenz
T
Eichhorst
B
Busch
R
et al
TP53 mutation and survival in chronic lymphocytic leukemia
J Clin Oncol
2010
28
29
4473
4479
24
Zenz
T
Kröber
A
Scherer
K
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
112
3322
3329
25
Dicker
F
Herholz
H
Schnittger
S
et al
The detection of TP53 mutations in chronic lymphocytic leukemia independently predicts rapid disease progression and is highly correlated with a complex aberrant karyotype
Leukemia
2009
23
117
124
26
Rossi
D
Cerri
M
Deambrogi
C
et al
The prognostic value of TP53 mutations in chronic lymphocytic leukemia is independent of Del17p13: implications for overall survival and chemorefractoriness
Clin Cancer Res
2009
15
995
1004
27
Malcikova
J
Smardova
J
Rocnova
L
et al
Monoallelic and biallelic inactivation of TP53 gene in chronic lymphocytic leukemia: selection, impact on survival, and response to DNA damage
Blood
2009
114
5307
5314
28
Zenz
T
Hoth
P
Busch
R
et al
TP53 Mutations and Outcome After Fludarabine and Cyclophosphamide (FC) or FC Plus Rituximab (FCR) in the CLL8 Trial of the GCLLSG [Abstract]
Blood
2009
114
22
1267
29
Gonzalez
D
Martinez
P
Wade
R
et al
Mutational Status of the TP53 Gene as a Predictor of Response and Survival in CLL Patients with and without 17p Deletion [Abstract]
Blood
2008
112
11
784
30
Stilgenbauer
S
Sander
S
Bullinger
L
et al
Clonal evolution in chronic lymphocytic leukemia: acquisition of high-risk genomic aberrations associated with unmutated VH, resistance to therapy, and short survival
Haematologica
2007
92
1242
1245
31
Shanafelt
TD
Witzig
TE
Fink
SR
et al
Prospective evaluation of clonal evolution during long-term follow-up of patients with untreated early-stage chronic lymphocytic leukemia
J Clin Oncol
2006
24
4634
4641
32
Zenz
T
Mohr
J
Eldering
E
et al
miR-34a as part of the resistance network in chronic lymphocytic leukemia
Blood
2009
113
3801
3808
33
Keating
MJ
Wierda
WG
Tam
CS
et al
Long Term Outcome Following Treatment Failure of FCR Chemoimmunotherapy as Initial Therapy for Chronic Lymphocytic Leukemia [Abstract]
Blood
2009
114
22
2381
34
Brown
CJ
Lain
S
Verma
CS
Fersht
AR
Lane
DP
Awakening guardian angels: drugging the p53 pathway
Nat Rev Cancer
2009
9
862
873
35
Reinhardt
HC
Jiang
H
Hemann
MT
Yaffe
MB
Exploiting synthetic lethal interactions for targeted cancer therapy
Cell Cycle
2009
8
3112
3119
36
Nahi
H
Lehmann
S
Mollgard
L
et al
Effects of PRIMA-1 on chronic lymphocytic leukaemia cells with and without hemizygous p53 deletion
Br J Haematol
2004
127
285
291
37
Pettitt
AR
Matutes
E
Oscier
D
Alemtuzumab in combination with high-dose methylprednisolone is a logical, feasible and highly active therapeutic regimen in chronic lymphocytic leukaemia patients with p53 defects
Leukemia
2006
20
1441
1445
38
Wierda
WG
Kipps
TJ
Mayer
J
et al
Ofatumumab as single-agent CD20 immunotherapy in fludarabine-refractory chronic lymphocytic leukemia
J Clin Oncol
2010
28
1749
1755
39
Keating
MJ
Flinn
I
Jain
V
et al
Therapeutic role of alemtuzumab (Campath-1H) in patients who have failed fludarabine: results of a large international study
Blood
2002
99
3554
3561
40
Fischer
K
Cramer
P
Stilgenbauer
S
et al
Bendamustine Combined with Rituximab (BR) in First-Line Therapy of Advanced CLL: A Multicenter Phase II Trial of the German CLL Study Group (GCLLSG) [Abstract]
Blood
2009
114
22
205
41
Bosch
F
Abrisqueta
P
Villamor
N
et al
Rituximab, fludarabine, cyclophosphamide, and mitoxantrone: a new, highly active chemoimmunotherapy regimen for chronic lymphocytic leukemia
J Clin Oncol
2009
27
4578
4584
42
Tsimberidou
AM
Keating
MJ
Treatment of fludarabine-refractory chronic lymphocytic leukemia
Cancer
2009
115
2824
2836
43
Stilgenbauer
S
Zenz
T
Winkler
D
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
27
3994
4001
44
Hillmen
P
Skotnicki
AB
Robak
T
et al
Alemtuzumab compared with chlorambucil as first-line therapy for chronic lymphocytic leukemia
J Clin Oncol
2007
25
5616
5623
45
Byrd
JC
Lin
TS
Dalton
JT
et al
Flavopiridol administered using a pharmacologically derived schedule is associated with marked clinical efficacy in refractory, genetically high-risk chronic lymphocytic leukemia
Blood
2007
109
399
404
46
Chanan-Khan
A
Porter
CW
Immunomodulating drugs for chronic lymphocytic leukaemia
Lancet Oncol
2006
7
480
488
47
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
23
2971
2979
48
Boettcher
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
23
2007
2017
49
Boettcher
S
Fischer
K
Stilgenbauer
S
et al
Quantitative MRD Assessments Predict Progression Free Survival in CLL Patients Treated with Fludarabine and Cyclophosphamide with or without Rituximab - a Prospective Analysis in 471 Patients from the Randomized GCLLSG CLL8 Trial [Abstract]
Blood
2008
112
11
326
50
Dreger
P
Corradini
P
Kimby
E
et al
Indications for allogeneic stem cell transplantation in chronic lymphocytic leukemia: the EBMT transplant consensus
Leukemia
2007
21
12
17
51
Dreger
P
Dohner
H
Ritgen
M
et al
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
116
2438
2447
52
Pettitt
AR
Matutes
E
Dearden
C
et al
Results of the phase II NCRI CLL206 trial of alemtuzumab in combination with high-dose methylprednisolone for high-risk (17P-) CLL
Haematologica
2009
94
1
694
53
Ferrajoli
A
Lee
BN
Schlette
EJ
et al
Lenalidomide induces complete and partial remissions in patients with relapsed and refractory chronic lymphocytic leukemia
Blood
2008
111
5291
5297
54
Chanan-Khan
A
Miller
KC
Musial
L
et al
Clinical efficacy of lenalidomide in patients with relapsed or refractory chronic lymphocytic leukemia: results of a phase II study
J Clin Oncol
2006
24
5343
5349
55
Lin
TS
Ruppert
AS
Johnson
AJ
et al
Phase II study of flavopiridol in relapsed chronic lymphocytic leukemia demonstrating high response rates in genetically high-risk disease
J Clin Oncol
2009
27
6012
6018
56
O'Brien
SM
Kantarjian
H
Thomas
DA
et al
Rituximab dose-escalation trial in chronic lymphocytic leukemia
J Clin Oncol
2001
19
2165
2170
57
Castro
JE
Sandoval-Sus
JD
Bole
J
et al
Rituximab in combination with high-dose methylprednisolone for the treatment of fludarabine refractory high-risk chronic lymphocytic leukemia
Leukemia
2008
22
2048
2053