For many years there has been considerable disassociation between the understood biology of chronic lymphocytic leukemia (CLL) and the therapeutics used to treat this disease. With the introduction of the first targeted CD20 antibody rituximab and its addition to chemotherapy came the first observation that minimal residual disease–negative (MRD-negative) complete responses (CRs) could be obtained with dramatically improved progression-free survival and overall survival. This advance was soon to be surpassed by the introduction of therapeutics that target B-cell receptor (BCR) signaling. New data show that BCR-inhibiting agents are very active for the treatment of relapsed CLL, despite the lack of MRD-negative CR, with durability of response being considerably more impressive than previously observed with other agents not producing MRD-negative CRs. This perspective provides a view of where these agents may take us in the future as CLL therapy evolves with this exciting new class of drugs.

Recognition that B-cell receptor (BCR) signaling is essential for the proliferation and survival of chronic lymphocytic leukemia (CLL) cells stands as one of the most important insights into the pathobiology of the disease. Accumulated evidence supports that antigen-dependent and -independent BCR signaling plays a central role in the pathogenesis of CLL (reviewed in Stevenson et al1  and Woyach et al2 ). Well-characterized molecular markers correlated with adverse prognosis, such as unmutated immunoglobulin heavy-chain chains3  and ZAP-704,5  expression, are now understood to be associated with and/or potentiate BCR-signaling activity, likely accounting for the more rapid progression of disease in cases where these features are present. Examination of CLL cells in the blood, bone marrow, and nodal compartment demonstrates that the BCR pathway is activated in the former two with enhanced proliferation of tumor cells.6  This matches the current concept of CLL expanding as a consequence of proliferation centers in the bone marrow, lymph nodes, and spleen. More recently, kinases immediately downstream of the BCR, including spleen tyrosine kinase (SYK) and phosphatidylinositol 3-kinase (PI3K), have been found to be constitutively activated in the majority of CLL patients.7-9  These kinases and downstream amplification kinases such as Bruton agammaglobulinemia tyrosine kinase (BTK) appear essential not only for activation of multiple survival pathways (Akt, Erk, nuclear factor κB) but also for chemokine-mediated migration and adhesion of B cells in the microenvironment.

Several small molecules have been developed to inhibit a variety of kinases in the BCR pathway, including LYN, SYK, BTK, and PI3K, with varied specificity. Pharmacologic inhibition of these kinases promotes apoptosis of CLL cells in vitro.9-12  After treatment with the SYK inhibitor fostamatinib,13  the first BCR-targeted agent to reach the clinic, rapid reduction in nodal volume, disease-related symptoms, and cytopenias was accompanied by a so-called “redistribution lymphocytosis.” This phenomenon is now recognized as a class effect of BCR antagonists, further supporting the role of BCR signaling in homing and retention of CLL cells within their supporting microenvironment and does not constitute progressive disease.14  The emergence of orally bioavailable, relatively nontoxic inhibitors of BCR-signaling kinases, particularly those directed at BTK and the p110δ PI3K isoform, represents not only a triumph of translational science but also a therapeutic advance of as yet undetermined clinical implications for CLL. As data emerge from clinical trials with these and other highly active therapies, clinicians caring for CLL patients are left with questions of how best to incorporate these agents into their treatment approaches.15  This article provides some insight on how these agents might alter future CLL therapy.

PI3K

Idelalisib (CAL-101, GS-1101) is a first-in-class, selective oral inhibitor of the p110δ isoform of PI3Kδ. Preclinical work with this molecule demonstrated that this small molecule inhibited both intrinsic and extrinsic survival signals, including those generated by BCR signaling in CLL,9,16,17  and prior studies of a PI3Kδ mutant mouse suggested predominately a B-cell phenotype, further supporting targeting this kinase.18  A phase 1 study that enrolled 54 patients with heavily pretreated relapsed/refractory CLL treated them with continuous once- or twice-daily doses ranging from 50 to 350 mg per dose.19  Responses, characterized by regression of lymphadenopathy and organomegaly and normalization of cytopenias, were observed within weeks of starting treatment (median, 1.9 months). After a median 9 months of drug exposure, an overall response rate (ORR) of 39% using the International Workshop on Chronic Lymphocytic Leukaemia (IWCLL) 2008 criteria was observed. Nodal response (>50% reduction from baseline) was observed in a larger proportion of patients (81%) who did not meet criteria for objective response, largely as a consequence of persisting peripheral blood lymphocytosis. Median progression-free survival (PFS) was 17 months; it increased to 29 months for those receiving 150 mg twice per day or greater. Dose-limiting toxicities were not observed, and potentially treatment-related adverse events (chiefly fatigue, rash, diarrhea, respiratory tract infections, and reversible increases in hepatic transaminases) resulted in discontinuation of treatment in only 7% of patients. Because PI3Kδ influences clonal expansion and differentiation of suppressor T cells, some of these events, particularly diarrhea and/or colitis, may represent on-target toxicities of idelalisib.20-22 

IPI-145 is the second PI3K-targeted agent to enter clinical development. IPI-145 potently inhibits both the p110δ and p110γ isoforms of the enzyme at pico- and nanomolar concentrations, respectively. Clinically, a lower dose of 25 mg twice daily predominately inhibits p110δ, while the maximally tolerated dose of 75 mg twice daily appears to inhibit both isoforms. Preliminary results of the phase 1 trial were recently presented.19  Relapsed/refractory CLL patients (N = 34; 22 evaluable for response) were treated with escalating doses from 8 to 100 mg twice per day; at the higher dose, 2 dose-limiting toxicities were reported (grade 3 rash, grade 3 elevation of hepatic transaminases). The median time to response (1.9 months) and pattern of response (ORR, 55%) was similar to the δ-specific agent. Similar to idelalisib, the nodal response rate was impressive (87%), and responses were observed without regard to genetic features or extent of prior therapy. However, the addition of p110γ inhibition at higher doses of IPI-145 appeared to result in more significant immune suppression; Pneumocystis pneumonia has been observed, and prophylaxis for opportunistic infections will be mandated going forward. Accrual continues to expansion cohorts enrolling both relapsed/refractory and treatment-naïve disease.

BTK

The first BTK inhibitor to enter the clinic was the orally bioavailable, irreversibly binding small molecule ibrutinib (PCI-32765).23  Mutation of BTK also occurs naturally in humans, resulting in a phenotype characterized by humoral insufficiency. Preclinical work with ibrutinib in CLL demonstrated inhibition of both intrinsic and extrinsic survival signals mediated by BTK.11,24,25  A phase 1 study of ibrutinib in B-cell malignancies was initiated in which durable clinical activity was noted in non-Hodgkin lymphoma and 9 of 16 CLL/small lymphocytic lymphoma patients.26  No dose-limiting toxicity was identified, and toxicity, including myelosuppression, was modest. In a recently published phase 1b study of ibrutinib, 85 patients with relapsed/refractory CLL/small lymphocytic lymphoma were enrolled at two different doses (420 or 840 mg daily).27  Extended therapy with ibrutinib was well tolerated, with common adverse events including grade 1 to 2 diarrhea, cough, fatigue, upper respiratory infections, nausea, fever, peripheral edema, myalgias, and petechiae/ecchymoses. Most adverse events with ibrutinib resolved despite continued treatment. Grade 3 or greater infections occurred more frequently early in therapy; the average rate per 100 patient-months within the first 6 months was 7.1 but 2.6 thereafter. After subarachnoid hemorrhages were reported in several patients receiving concomitant warfarin treatment, but not other anticoagulants or antiplatelet agents, concurrent therapy with oral vitamin K antagonists has been prohibited. Further, the manufacturer has provided guidance for holding the drug around the time of invasive procedures. BTK does not appear essential for platelet activation, but a role for BTK inhibition in stable thrombus formulation has been postulated.28  Yet despite the fact that platelets from patients with X-linked agammmaglobulinemia demonstrate abnormalities of collagen and collagen-related peptide-induced aggregation,29  bleeding diatheses are not observed clinically.30  Ongoing randomized trials will help better characterize the potential risk for significant bleeding complications attributable to pharmacologic BTK inhibition.

Like other BCR antagonists, an early increase in lymphocytosis is typically noted by day 7 and persists for 2 to 3 months before slowly declining over time, concomitant with notable reduction in lymph node and spleen size and improvement in cytopenias. The ORR by IWCLL 2008 criteria31  was 71% (2 complete responses [CRs], 34 partial responses) in the 420-mg cohort and 71% (24 partial responses) in the 840-mg cohort. In addition, 10 (20%) and 5 patients (15%) in the 420-mg and 840-mg cohorts, respectively, demonstrated a nodal response with persistent lymphocytosis. Response to ibrutinib did not vary on the basis of any adverse feature previously identified in CLL. Most notably, of the 28 patients with del(17p) enrolled on this study, 18 (68%) responded. The 26-month estimated PFS for all patients enrolled on this study was 75%. Unlike response, PFS did differ by genomic group; 26-month estimated PFS was 56% for patients with either del(11q22.3) or del(17p13.1) vs 93% in patients without either of these abnormalities. Collectively, these suggest that ibrutinib remissions are quite durable compared with other therapies used in this setting.

There can be no question that the widespread availability of BCR-signaling antagonists will rapidly alter the nature of CLL therapy (Table 1). How they will be used is in part dependent upon the labeled indications, unknown until regulatory agency approval, as well as the outcomes of several ongoing registration studies (Table 2). Nonetheless, the efficacy, durability of remission, and safety profile of ibrutinib for most patients will likely lead to rapid adoption in the salvage setting. And while available data suggest that the efficacy, durability of remission, and safety profile of single-agent idelalisib may be less impressive compared with ibrutinib, that drug also appears to represent a significant advance over many options CLL patients today receive in the setting of relapse. Idelalisib application might therefore be narrowed initially to patients not appropriate for ibrutinib, such as those on warfarin or those who are intolerant of the drug. However, beyond merely replacing older agents in the routine management of CLL, these new agents raise more fundamental questions about the changing natural history of the disease, goals of treatment, and even the prospect of curative therapy. We raise several of these as yet unanswered questions below and provide perspective on each.

Table 1

Active registration studies for BCR antagonists

AgentStudy title/numberClinicaltrials.gov identifierPhaseIndicationDesign
Ibrutinib Resonate/PCYC-1112-CA NCT01578707 Relapsed/refractory Ofatumumab vs ibrutinib 
 Resonate-2/PCYC-1115-CA NCT01722487 Treatment-naïve Chlorambucil vs ibrutinib 
 Resonate-17/PCYC-1117-CA NCT01744691 Relapsed/refractory del(17p)+ Ibrutinib 
Idelalisib GS-US-312-0115 NCT01569295 Relapsed/refractory Bendamustine, rituximab ± idelalisib 
 GS-US-312-0116 NCT01539512 Relapsed/refractory Rituximab ± idelalisib 
 GS-US-312-0119 NCT01659021 Relapsed/refractory Ofatumumab ± idelalisib 
AgentStudy title/numberClinicaltrials.gov identifierPhaseIndicationDesign
Ibrutinib Resonate/PCYC-1112-CA NCT01578707 Relapsed/refractory Ofatumumab vs ibrutinib 
 Resonate-2/PCYC-1115-CA NCT01722487 Treatment-naïve Chlorambucil vs ibrutinib 
 Resonate-17/PCYC-1117-CA NCT01744691 Relapsed/refractory del(17p)+ Ibrutinib 
Idelalisib GS-US-312-0115 NCT01569295 Relapsed/refractory Bendamustine, rituximab ± idelalisib 
 GS-US-312-0116 NCT01539512 Relapsed/refractory Rituximab ± idelalisib 
 GS-US-312-0119 NCT01659021 Relapsed/refractory Ofatumumab ± idelalisib 
Table 2

Selected BCR-signaling antagonists in clinical development for CLL indications

TargetAgentManufacturerStudy phase
BTK Ibrutinib (PCI-32765) Pharmacyclics/Janssen 
 CC-292 (AVL-292) Celgene 1b/2 
 GDC-0834 Genentech 
 ACP-196 Acerta 
 ONO-WG-307 Ono 
PI3Kδ Idelalisib (GS-1101, CAL-101) Gilead Sciences 
 GS-9820 Gilead Sciences 
 AMG-319 Amgen 
 TGR-1202 TG Therapeutics 
PI3Kδ/γ IPI-145 Infinity 
Syk GS-9973 Gilead Sciences 
TargetAgentManufacturerStudy phase
BTK Ibrutinib (PCI-32765) Pharmacyclics/Janssen 
 CC-292 (AVL-292) Celgene 1b/2 
 GDC-0834 Genentech 
 ACP-196 Acerta 
 ONO-WG-307 Ono 
PI3Kδ Idelalisib (GS-1101, CAL-101) Gilead Sciences 
 GS-9820 Gilead Sciences 
 AMG-319 Amgen 
 TGR-1202 TG Therapeutics 
PI3Kδ/γ IPI-145 Infinity 
Syk GS-9973 Gilead Sciences 

Are “remission” and “ disease control” still synonymous?

Long-term disease control of CLL in the chemoimmunotherapy era has strongly correlated with quality and depth of remission.32  Achievement of minimal residual disease–negative (MRD-negative) CR after fludarabine-based chemoimmunotherapy has predicted not only longer time to treatment failure but also prolonged PFS and overall survival (OS).32  Failure to achieve at least a PR after fludarabine, cyclophosphamide, and rituximab therapy generally bodes a dismal prognosis, particularly among patients with genetically high-risk disease.33  On the other hand, most patients treated with BCR-signaling antagonists ultimately achieve durable but no better than partial remissions, and CRs have rarely been observed after single-agent therapy. Although data are limited, bone marrow biopsies typically demonstrate persistence of disease that slowly diminishes over time, even when peripheral blood lymphocytosis and lymphadenopathy are resolved.

Strikingly, long-term remissions are observed, even among patients who never achieved resolution of peripheral blood lymphocytosis. Duration of response, then, may not necessarily correlate with depth of response after kinase inhibitor therapy, and remission may not be necessary to effect durable clinical benefit, provided therapy is continued. The ability to continue BCR-directed therapy for an extended period of time differentiates it from chemotherapy approaches in which this is generally prohibitively toxic. These observations have already prompted reassessment of consensus criteria for clinical trial outcomes,14,31  but they might necessarily prompt a more general re-examination of the end points for CLL therapy. Intermediate end points such as response, appropriate for a relapsing/remitting natural history, may ultimately prove inappropriate when the disease is chronically controlled and the treatment is continuous rather than episodic. Freedom from disease-related complications (infection, transfusion, autoimmune phenomenon) and survival end points will likely emerge as more meaningful outcome measures. CLL investigators will undoubtedly need to develop new intermediate end points predictive of disease outcome that can be efficiently incorporated into clinical trial designs.

On the other hand, enthusiasm for long-term maintenance therapy with drugs, which as single agents promote durable disease control without achievement of MRD-negative remission, must be tempered by related concerns for patient adherence and cost. Here the imatinib experience is informative. Adherence to kinase inhibitor therapy has been associated with the achievement of important treatment end points in chronic myelogenous leukemia such as major molecular response.34  However, factors influencing adherence to oral cancer therapies are poorly understood, and validated strategies to promote compliance are likewise limited.35  There is increasing concern that economic hardship could limit patients’ abilities not only to elect but also to adhere to newer, more effective therapies.36  Although patients’ collective compliance with prescribed medications may be cost-saving from a health system perspective, out-of-pocket costs to individual patients for ever more costly therapies remains an unresolved issue.37,38  While the costs of these newer BCR kinase inhibitors are as yet a matter of speculation, clinicians are rightly concerned for the price tag of innovation.39 

Do accepted genetic risk models still hold in the kinase inhibitor era?

Treatment guidelines currently recommend that patients with short remissions after chemoimmunotherapy or those with del(17p13.1) consider aggressive therapeutic interventions, including reduced-intensity allogeneic stem cell transplantation, as part of initial therapy.40,41  Although the experience with BCR inhibitors is relatively immature to date, patients with del(17p13.1) demonstrated 57% PFS at 26 months in the initial report of ibrutinib, irrespective of the number of prior of treatments.27  In general, patients on these agents feel well and often elect to defer this stem cell transplantation if possible. As ibrutinib data have matured, our own practice has been to counsel patients about all available data but to strongly recommend allogeneic transplantation only to CLL patients who either responded poorly to ibrutinib or who lacked subsequent cytoreductive options that could render transplantation impossible in the event of ibrutinib breakthrough. As genetic mechanisms of resistance to ibrutinib become better characterized, sensitive monitoring techniques for their emergence might also prompt this recommendation.

Perhaps the more pressing concern as BCR inhibitors approach widespread application in the clinic is a more complete understanding of the molecular and genetic factors underlying and predicting for drug resistance. Little data has yet been presented regarding potential mechanisms of idelalisib resistance, and the first report describing mechanisms of resistance among ibrutinib-treated CLL patients has only recently been presented. In the latter case, treatment-emergent single nucleotide variations encoding a cysteine-to-serine substitution at position 481 of BTK (C481S), impairing covalent binding of ibrutinib to BTK, and a potential gain-of-function mutation (R665W) substitution in PLCλ2, have been identified.42  Pretreatment factors predicting emergence of resistance are poorly characterized, although the majority of treatment failures appear to occur among patients with adverse genetic risk features such as complex karyotype, del(11q22.3), and del(17p13.1). The mechanism by which this occurs is uncertain at this time. Whether BCR-inhibitor therapy will influence the natural history of relapsed disease (ie, emergence of Richter’s transformation, clonal evolution) is an important question to which emerging data from randomized studies will hopefully provide a rapid answer.

What is the rationale for combination therapy and which combinations are rational?

Initial trials of both ibrutinib- and idelalisib-based combination therapies have also been conducted. Early results from a phase 1b/2 study of ibrutinib in combination with the standard dose and schedule of ofatumumab have been presented,43  and a more recent phase 2 study exploring conventional doses of rituximab in combination with ibrutinib is ongoing.44  In both studies, redistribution lymphocytosis appears to be attenuated, peaking earlier and resolving more rapidly. Responses have been observed across all commonly accepted genetic risk groups, including del(17p13.1).43,44  Encouraging responses were likewise observed when patients were treated with standard-dose bendamustine and rituximab in combination with ibrutinib.45  The ORR of the combination was 93%, including 71% (14% CR) in the del(17p) subset. Notably, prior studies have failed to report CRs after bendamustine and rituximab treatment of del(17p) CLL. The addition of idelalisib to standard doses and schedules of rituximab, bendamustine, or the combination has also been explored.46  Response rates (ORR, 81%) and time to response (median, 1.9 months) were similar in all 3 arms of the study. At 2 years of follow-up, 71% of responses persisted, and PFS and OS are estimated at 62% and 85%, respectively. Response rates were again similar across all disease subgroups, including those with bulky or genetic high-risk disease.

These outcomes are informative for future combination studies. First, it is not yet clear that the combination therapies assessed to date represent a significant advance over the single agents. Combination studies reporting higher CR rates have not yet reported more durable disease control. Most notable may be the suggestion that chemotherapy or chemoimmunotherapy does not result in significantly higher ORRs than the combinations that do not include cytotoxic agents. And while the addition of rituximab to cytotoxic chemotherapy has been convincingly demonstrated to improve survival in CLL,47  it will again require randomized trials to rigorously explore potential gains in survival when kinase inhibitors are considered. The lack of added dramatic benefit of CD20 antibody to ibrutinib could in part relate to its recently recognized effects on interleukin-2–induced T-cell kinase (ITK).48  An unintended effect of ibrutinib in this setting could be inhibition of natural killer cell–mediated antibody-dependent cellular cytotoxicity that is dependent on functional ITK49  which could limit CD20-directed antibody efficacy. Hence, our previous principles of just adding therapeutic antibodies or other immune therapies to these BCR-signaling agents will have to be considered carefully in the context of influence on other components of the immune system.

The most exciting outcomes will likely result when the principles of combination chemotherapy are reimagined for a new era and mechanistically distinct targeted agents with nonoverlapping toxicities are combined. For instance, combinations of BCR-signaling antagonists targeting different kinases in the same pathway could optimize inhibition of BCR signaling and potentially prevent emergence of resistance. Alternatively, an equally attractive approach recapitulates in reverse the genetic experiment showing BCL2 rescues BTK-deficient B lymphocytes in normal murine B cells.50  CLL is a disease in which bcl-2 is overexpressed51  that also appears to be influenced by BTK inhibition. ABT-199, a small molecule inhibitor of BCL2, might be rationally combined with ibrutinib in this setting, given the promising results emerging from phase 1 studies.52  Combinations using immunomodulatory agents also have strong preclinical rationale. For instance, lenalidomide has been shown to inhibit T-regulatory T cells,53  polarize Th2 T cells to a Th1 phenotype,54  and enhance natural killer cell function. In CLL55  patients, these immune-modulating properties of lenalidomide effects T-cell immune synapse repair,56,57  observed even when lenalidomide is given at relatively low doses. Ibrutinib, via ITK-mediated inhibition of Th2 T cells,48  has complementary immune-modulating potential. Combination therapy to target immune reconstitution in CLL remains appealing. Such pairings might also limit the toxicity of combination therapy, mindful that drug-drug interactions mediated by cytochrome P450 enzymes may be a consideration in combining many of these agents. For instance, sequential administration of kinase inhibitors might also attenuate the risk for tumor flare induced by lenalidomide. Clinical trials with these combinations are of great interest.

Can initial treatment with biologically based therapy change the natural history of disease?

There are as yet limited data with BCR-signaling antagonists for previously untreated, symptomatic CLL.58  A study of 31 patients age 65 years or older who were treated with ibrutinib has been preliminarily reported. The median age was 71 years, and more than half had advanced Rai stage disease. Only 9% of patients had high-risk genomic features, either del(11q22.3) or del(17p13.1). The ORR by IWCLL criteria was 71%, and an additional 13% achieved PR with lymphocytosis. At median 24 months of follow-up, PFS and OS were 96%. Toxicity was similar to that observed in patients treated for relapsed disease. These results are quite remarkable but only consider predominately low-risk genomic patients. Wiestner et al at the National Heart, Lung, and Blood Institute have reported a small cohort of 15 previously untreated del(17p13.1) patients in whom PFS was 87% at 15 months.59  Data with idelalisib are available in the same patient population but only when idelalisib is combined with rituximab. A phase 1b study included 64 elderly treatment-naïve patients treated with 8 weekly doses of rituximab in combination with idelalisib administered continuously at 150 mg twice per day for 48 weeks, after which time responding patients could continue idelalisib maintenance.60  Outcomes presented for the first 50 patients included an ORR of 97% (19% CR). PFS at 24 months was 93%, including all 9 patients with poor-prognosis TP53 mutation or del(17p).

The availability of these highly active BCR-signaling agents certainly merits their incorporation into investigational frontline regimens, such as the ongoing Resonate-2 study comparing ibrutinib to chlorambucil and the forthcoming Alliance A041202 study of untreated CLL patients age 65 and older, which will include 3 arms (bendamustine + rituximab, ibrutinib + rituximab, or ibrutinib alone). These trials essentially seek to eliminate chemotherapy from the initial treatment approach for elderly patients with CLL. Another approach particularly adaptable to young patients (age <65-70 years) will address the question of adding BCR-signaling agents to highly effective chemoimmunotherapy in hopes of affecting MRD-negative disease and potentially cure of CLL patients.

Can early intervention with biologically based therapy change the natural history of disease?

Newly diagnosed CLL patients learn that the disease is highly treatable but ultimately incurable. Early intervention with cytotoxic chemotherapy failed to alter the natural history of disease,61  and results of trials using chemoimmunotherapy in that context have not yet been reported. Consensus treatment guidelines for CLL therefore recommend that therapy be withheld until symptoms intervene or the disease progresses to an essentially arbitrary measure of burden.31  Consequently, much CLL therapy is directed at the alleviation of rather than the prevention of morbidity. But progressive declines in immune function are measurable long before treatment is indicated, and most existing therapies not only fail to reverse that damage but in many cases exacerbate the problem.

An overall reconsideration of the relative risks and merits of early intervention for asymptomatic disease is also now warranted. Particularly when high-risk cytogenetic features are present, a rationale for early intervention persists, since these remain the patients most likely to relapse during BCR kinase–directed therapy. Yet the favorable toxicity profile suggests that we might also upend the conventional wisdom of deferring therapy in elderly populations. Earlier intervention in elderly patients with low-grade lymphoma has been advocated as a strategy to prevent progression to more advanced disease when therapy is less well tolerated. We might then consider clinical trials investigating early intervention in elderly CLL to prevent the morbid complications of progressive disease and immune dysfunction among a patient population ill-equipped to tolerate the most effective conventional therapies. Trials designed to rigorously assess the clinical benefits of treatment, broadly understood, might substantially improve the lot of the average CLL patient often excluded from clinical trials. Not only the BCR-signaling agents but also immune restorative agents such as lenalidomide might be considered in this context.

This work was supported by the National Institutes of Health, National Cancer Institute (P01 CA95426, P50CA140158, and R01 CA177292), Four Winds Foundation, The Leukemia and Lymphoma Society, Harry Mangurian Foundation, Mr. and Mrs. Michael Thomas, and The D. Warren Brown Foundation.

Contribution: J.A.J. and J.C.B. wrote the paper and approved the final version.

Conflict-of-interest disclosure: J.C.B. was a consultant for Calistoga and receives milestone payments that have been contractually committed to an independent charity. J.A.J. declares no competing financial interests.

Correspondence: Jeffrey Jones, The Ohio State University, A350B Starling Loving Hall, 320 West 10th Ave, Columbus, OH 43210; e-mail: jeffrey.jones@osumc.edu; and John C. Byrd, The Ohio State University, CCC Bldg Room 455B, 410 West 12th Ave, Columbus, OH 43210; e-mail: john.byrd@osumc.edu.

1
Stevenson
 
FK
Krysov
 
S
Davies
 
AJ
Steele
 
AJ
Packham
 
G
B-cell receptor signaling in chronic lymphocytic leukemia.
Blood
2011
, vol. 
118
 
16
(pg. 
4313
-
4320
)
2
Woyach
 
JA
Johnson
 
AJ
Byrd
 
JC
The B-cell receptor signaling pathway as a therapeutic target in CLL.
Blood
2012
, vol. 
120
 
6
(pg. 
1175
-
1184
)
3
Rosenwald
 
A
Alizadeh
 
AA
Widhopf
 
G
, et al. 
Relation of gene expression phenotype to immunoglobulin mutation genotype in B cell chronic lymphocytic leukemia.
J Exp Med
2001
, vol. 
194
 
11
(pg. 
1639
-
1647
)
4
Wiestner
 
A
Rosenwald
 
A
Barry
 
TS
, et al. 
ZAP-70 expression identifies a chronic lymphocytic leukemia subtype with unmutated immunoglobulin genes, inferior clinical outcome, and distinct gene expression profile.
Blood
2003
, vol. 
101
 
12
(pg. 
4944
-
4951
)
5
Chen
 
L
Huynh
 
L
Apgar
 
J
, et al. 
ZAP-70 enhances IgM signaling independent of its kinase activity in chronic lymphocytic leukemia.
Blood
2008
, vol. 
111
 
5
(pg. 
2685
-
2692
)
6
Herishanu
 
Y
Pérez-Galán
 
P
Liu
 
D
, et al. 
The lymph node microenvironment promotes B-cell receptor signaling, NF-kappaB activation, and tumor proliferation in chronic lymphocytic leukemia.
Blood
2011
, vol. 
117
 
2
(pg. 
563
-
574
)
7
Buchner
 
M
Fuchs
 
S
Prinz
 
G
, et al. 
Spleen tyrosine kinase is overexpressed and represents a potential therapeutic target in chronic lymphocytic leukemia.
Cancer Res
2009
, vol. 
69
 
13
(pg. 
5424
-
5432
)
8
Baudot
 
AD
Jeandel
 
PY
Mouska
 
X
, et al. 
The tyrosine kinase Syk regulates the survival of chronic lymphocytic leukemia B cells through PKCdelta and proteasome-dependent regulation of Mcl-1 expression.
Oncogene
2009
, vol. 
28
 
37
(pg. 
3261
-
3273
)
9
Herman
 
SE
Gordon
 
AL
Wagner
 
AJ
, et al. 
Phosphatidylinositol 3-kinase-δ inhibitor CAL-101 shows promising preclinical activity in chronic lymphocytic leukemia by antagonizing intrinsic and extrinsic cellular survival signals.
Blood
2010
, vol. 
116
 
12
(pg. 
2078
-
2088
)
10
ten Hacken
 
E
Scielzo
 
C
Bertilaccio
 
MT
, et al. 
Targeting the LYN/HS1 signaling axis in chronic lymphocytic leukemia.
Blood
2013
, vol. 
121
 
12
(pg. 
2264
-
2273
)
11
Herman
 
SE
Gordon
 
AL
Hertlein
 
E
, et al. 
Bruton tyrosine kinase represents a promising therapeutic target for treatment of chronic lymphocytic leukemia and is effectively targeted by PCI-32765.
Blood
2011
, vol. 
117
 
23
(pg. 
6287
-
6296
)
12
Milara
 
J
Martinez-Losa
 
M
Sanz
 
C
, et al. 
 
Bafetinib inhibits functional responses of human eosinophils in vitro. Eur J Pharmacol. 2013;715(1-3):172-180
13
Friedberg
 
JW
Sharman
 
J
Sweetenham
 
J
, et al. 
Inhibition of Syk with fostamatinib disodium has significant clinical activity in non-Hodgkin lymphoma and chronic lymphocytic leukemia.
Blood
2010
, vol. 
115
 
13
(pg. 
2578
-
2585
)
14
Cheson
 
BD
Byrd
 
JC
Rai
 
KR
, et al. 
Novel targeted agents and the need to refine clinical end points in chronic lymphocytic leukemia.
J Clin Oncol
2012
, vol. 
30
 
23
(pg. 
2820
-
2822
)
15
Hallek
 
M
Signaling the end of chronic lymphocytic leukemia: new frontline treatment strategies.
Blood
 
2013;122(23):3723-3734
16
Hoellenriegel
 
J
Meadows
 
SA
Sivina
 
M
, et al. 
The phosphoinositide 3′-kinase delta inhibitor, CAL-101, inhibits B-cell receptor signaling and chemokine networks in chronic lymphocytic leukemia.
Blood
2011
, vol. 
118
 
13
(pg. 
3603
-
3612
)
17
Lannutti
 
BJ
Meadows
 
SA
Herman
 
SE
, et al. 
CAL-101, a p110delta selective phosphatidylinositol-3-kinase inhibitor for the treatment of B-cell malignancies, inhibits PI3K signaling and cellular viability.
Blood
2011
, vol. 
117
 
2
(pg. 
591
-
594
)
18
Okkenhaug
 
K
Bilancio
 
A
Farjot
 
G
, et al. 
Impaired B and T cell antigen receptor signaling in p110delta PI 3-kinase mutant mice.
Science
2002
, vol. 
297
 
5583
(pg. 
1031
-
1034
)
19
Brown
 
JR
Furman
 
RR
Flinn
 
I
, et al. 
 
Final results of a phase I study of idelalisib (GS-1101) a selective inhibitor of PI3Kδ, in patients with relapsed or refractory CLL [abstract]. J Clin Oncol. 2013;31(15). Abstract 7003
20
Uno
 
JK
Rao
 
KN
Matsuoka
 
K
, et al. 
 
Altered macrophage function contributes to colitis in mice defective in the phosphoinositide-3 kinase subunit p110δ. Gastroenterology. 2010;139(5):1642-1653
21
Patton
 
DT
Garden
 
OA
Pearce
 
WP
, et al. 
Cutting edge: the phosphoinositide 3-kinase p110 delta is critical for the function of CD4+CD25+Foxp3+ regulatory T cells.
J Immunol
2006
, vol. 
177
 
10
(pg. 
6598
-
6602
)
22
Okkenhaug
 
K
Patton
 
DT
Bilancio
 
A
Garçon
 
F
Rowan
 
WC
Vanhaesebroeck
 
B
The p110delta isoform of phosphoinositide 3-kinase controls clonal expansion and differentiation of Th cells.
J Immunol
2006
, vol. 
177
 
8
(pg. 
5122
-
5128
)
23
Honigberg
 
LA
Smith
 
AM
Sirisawad
 
M
, et al. 
The Bruton tyrosine kinase inhibitor PCI-32765 blocks B-cell activation and is efficacious in models of autoimmune disease and B-cell malignancy.
Proc Natl Acad Sci U S A
2010
, vol. 
107
 
29
(pg. 
13075
-
13080
)
24
Ponader
 
S
Chen
 
SS
Buggy
 
JJ
, et al. 
The Bruton tyrosine kinase inhibitor PCI-32765 thwarts chronic lymphocytic leukemia cell survival and tissue homing in vitro and in vivo.
Blood
2012
, vol. 
119
 
5
(pg. 
1182
-
1189
)
25
de Rooij
 
MF
Kuil
 
A
Geest
 
CR
, et al. 
The clinically active BTK inhibitor PCI-32765 targets B-cell receptor- and chemokine-controlled adhesion and migration in chronic lymphocytic leukemia.
Blood
2012
, vol. 
119
 
11
(pg. 
2590
-
2594
)
26
Advani
 
RH
Buggy
 
JJ
Sharman
 
JP
, et al. 
Bruton tyrosine kinase inhibitor ibrutinib (PCI-32765) has significant activity in patients with relapsed/refractory B-cell malignancies.
J Clin Oncol
2013
, vol. 
31
 
1
(pg. 
88
-
94
)
27
Byrd
 
JC
Furman
 
RR
Coutre
 
SE
, et al. 
Targeting BTK with ibrutinib in relapsed chronic lymphocytic leukemia.
N Engl J Med
2013
, vol. 
369
 
1
(pg. 
32
-
42
)
28
Liu
 
J
Fitzgerald
 
ME
Berndt
 
MC
Jackson
 
CW
Gartner
 
TK
Bruton tyrosine kinase is essential for botrocetin/VWF-induced signaling and GPIb-dependent thrombus formation in vivo.
Blood
2006
, vol. 
108
 
8
(pg. 
2596
-
2603
)
29
Quek
 
LS
Bolen
 
J
Watson
 
SP
A role for Bruton’s tyrosine kinase (Btk) in platelet activation by collagen.
Curr Biol
1998
, vol. 
8
 
20
(pg. 
1137
-
1140
)
30
Futatani
 
T
Watanabe
 
C
Baba
 
Y
Tsukada
 
S
Ochs
 
HD
Bruton’s tyrosine kinase is present in normal platelets and its absence identifies patients with X-linked agammaglobulinaemia and carrier females.
Br J Haematol
2001
, vol. 
114
 
1
(pg. 
141
-
149
)
31
Hallek
 
M
Cheson
 
BD
Catovsky
 
D
, et al. 
International Workshop on Chronic Lymphocytic Leukemia
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
, vol. 
111
 
12
(pg. 
5446
-
5456
)
32
Böttcher
 
S
Ritgen
 
M
Fischer
 
K
, et al. 
Minimal residual disease quantification is an independent predictor of progression-free and overall survival in chronic lymphocytic leukemia: a multivariate analysis from the randomized GCLLSG CLL8 trial.
J Clin Oncol
2012
, vol. 
30
 
9
(pg. 
980
-
988
)
33
Tam
 
CS
O’Brien
 
S
Wierda
 
W
, et al. 
Long-term results of the fludarabine, cyclophosphamide, and rituximab regimen as initial therapy of chronic lymphocytic leukemia.
Blood
2008
, vol. 
112
 
4
(pg. 
975
-
980
)
34
Marin
 
D
Bazeos
 
A
Mahon
 
FX
, et al. 
 
Adherence is the critical factor for achieving molecular responses in patients with chronic myeloid leukemia who achieve complete cytogenetic responses on imatinib. J Clin Oncol. 2010;28(14):2381-2388
35
Efficace
 
F
Baccarani
 
M
Rosti
 
G
, et al. 
 
Investigating factors associated with adherence behaviour in patients with chronic myeloid leukemia: an observational patient-centered outcome study. Br J Cancer. 2012;107(6):904-909
36
Kelley
 
RK
Venook
 
AP
 
Nonadherence to imatinib during an economic downturn. N Engl J Med. 2010;363(6):596-598
37
Wu
 
EQ
Johnson
 
S
Beaulieu
 
N
, et al. 
 
Healthcare resource utilization and costs associated with non-adherence to imatinib treatment in chronic myeloid leukemia patients. Curr Med Res Opin. 2010;26(1):61-69
38
Darkow
 
T
Henk
 
HJ
Thomas
 
SK
, et al. 
Treatment interruptions and non-adherence with imatinib and associated healthcare costs: a retrospective analysis among managed care patients with chronic myelogenous leukaemia.
Pharmacoeconomics
2007
, vol. 
25
 
6
(pg. 
481
-
496
)
39
Zhang
 
W
Trachootham
 
D
Liu
 
J
, et al. 
 
Stromal control of cystine metabolism promotes cancer cell survival in chronic lymphocytic leukaemia. Nat Cell Biol. 2012;14(3):276-286
40
Zelenetz
 
AD
Wierda
 
WG
Abramson
 
JS
, et al. 
National Comprehensive Cancer Network
Non-Hodgkin’s lymphomas, version 1.2013.
J Natl Compr Canc Netw
2013
, vol. 
11
 
3
(pg. 
257
-
272
)
41
Pospisilova
 
S
Gonzalez
 
D
Malcikova
 
J
, et al. 
ERIC recommendations on TP53 mutation analysis in chronic lymphocytic leukemia.
Leukemia
 
2012;26(7):1458-1461
42
Chang
 
BY
Furman
 
RR
Zapatka
 
M
, et al. 
 
Use of tumor genomic profiling to reveal mechanisms of resistance to the BTK inhibitor ibrutinib in chronic lymphocytic leukemia (CLL) [abstract]. J Clin Oncol. 2013;31(15). Abstract 7014
43
Jaglowski
 
SM
Jones
 
JA
Flynn
 
JM
, et al. 
 
A phase Ib/II study evaluating activity and tolerability of BTK inhibitor PCI-32765 and ofatumumab in patients with chronic lymphocytic leukemia/small lymphocytic lymphoma (CLL/SLL) and related diseases [abstract]. J Clin Oncol. 2012;30(15). Abstract 6508
44
Burger
 
JA
Keating
 
MJ
Wierda
 
WG
, et al. 
The Btk inhibitor ibrutinib (PCI-32765) in combination with rituximab is well tolerated and displays profound activity in high-risk chronic lymphocytic leukemia (CLL) patients [abstract].
Blood
2012
, vol. 
120
  
(21). Abstract 187
45
O’Brien
 
SM
Barrientos
 
JC
Flinn
 
IW
, et al. 
 
Combination of the Bruton’s tyrosine kinase (BTK) inhibitor PCI-32765 with bendamustine (B)/rituximab (R) (BR) in patients (pts) with relapsed/refractory (R/R) chronic lymphocytic leukemia (CLL): Interim results of a phase Ib/II study [abstract]. J Clin Oncol. 2012;30(15). Abstract 6515
46
Barrientos
 
JC
Furman
 
RR
Leonard
 
J
, et al. 
 
Update on a phase I study of the selective PI3Kδ inhibitor idelalisib (GS-1101) in combination with rituximab and/or bendamustine in patients with relapsed or refractory CLL [abstract]. J Clin Oncol. 2013;31(15). Abstract 7017
47
Hallek
 
M
Fischer
 
K
Fingerle-Rowson
 
G
, et al. 
International Group of Investigators; German Chronic Lymphocytic Leukaemia Study Group
Addition of rituximab to fludarabine and cyclophosphamide in patients with chronic lymphocytic leukaemia: a randomised, open-label, phase 3 trial.
Lancet
2010
, vol. 
376
 
9747
(pg. 
1164
-
1174
)
48
Dubovsky
 
JA
Beckwith
 
KA
Natarajan
 
G
, et al. 
Ibrutinib is an irreversible molecular inhibitor of ITK driving a Th1-selective pressure in T lymphocytes.
Blood
2013
, vol. 
122
 
15
(pg. 
2539
-
2549
)
49
Khurana
 
D
Arneson
 
LN
Schoon
 
RA
Dick
 
CJ
Leibson
 
PJ
Differential regulation of human NK cell-mediated cytotoxicity by the tyrosine kinase Itk.
J Immunol
2007
, vol. 
178
 
6
(pg. 
3575
-
3582
)
50
Woodland
 
RT
Schmidt
 
MR
Korsmeyer
 
SJ
Gravel
 
KA
Regulation of B cell survival in xid mice by the proto-oncogene bcl-2.
J Immunol
1996
, vol. 
156
 
6
(pg. 
2143
-
2154
)
51
Kitada
 
S
Andersen
 
J
Akar
 
S
, et al. 
Expression of apoptosis-regulating proteins in chronic lymphocytic leukemia: correlations with in vitro and in vivo chemoresponses.
Blood
1998
, vol. 
91
 
9
(pg. 
3379
-
3389
)
52
Seymour
 
JF
Davids
 
MS
Pagel
 
JM
, et al. 
 
Updated results of a phase I first-in-human study of the BCL-2 inhibitor ABT-199 (GDC-0199) in patients with relapsed/refractory (R/R) chronic lymphocytic leukemia (CLL) [abstract]. J Clin Oncol. 2013;31(15). Abstract 7018
53
Wobus
 
M
Benath
 
G
Ferrer
 
RA
, et al. 
 
Impact of lenalidomide on the functional properties of human mesenchymal stromal cells. Exp Hematol. 2012;40(10):867-876
54
Xu
 
W
Celeridad
 
M
Sankar
 
S
Webb
 
DR
Bennett
 
BL
CC-4047 promotes Th1 cell differentiation and reprograms polarized human Th2 cells by enhancing transcription factor T-bet.
Clin Immunol
2008
, vol. 
128
 
3
(pg. 
392
-
399
)
55
Wu
 
L
Adams
 
M
Carter
 
T
, et al. 
Lenalidomide enhances natural killer cell and monocyte-mediated antibody-dependent cellular cytotoxicity of rituximab-treated CD20+ tumor cells.
Clin Cancer Res
2008
, vol. 
14
 
14
(pg. 
4650
-
4657
)
56
Ramsay
 
AG
Johnson
 
AJ
Lee
 
AM
, et al. 
Chronic lymphocytic leukemia T cells show impaired immunological synapse formation that can be reversed with an immunomodulating drug.
J Clin Invest
2008
, vol. 
118
 
7
(pg. 
2427
-
2437
)
57
Shanafelt
 
TD
Ramsay
 
AG
Zent
 
CS
, et al. 
Long-term repair of T-cell synapse activity in a phase II trial of chemoimmunotherapy followed by lenalidomide consolidation in previously untreated chronic lymphocytic leukemia (CLL).
Blood
2013
, vol. 
121
 
20
(pg. 
4137
-
4141
)
58
O’Brien
 
S
Furman
 
RR
Coutre
 
SE
, et al. 
Ibrutinib as initial therapy for elderly patients with chronic lymphocytic leukaemia or small lymphocytic lymphoma: an open-label, multicentre, phase 1b/2 trial.
Lancet Oncol
2014
 
15(1)48-58
59
Wiestner
 
A
Herman
 
S
Mustafa
 
R
, et al. 
 
Potent single agent activity of Ibrutinib (PCI-32765) in patients with chronic lymphocytic leukemia (CLL): clinical and translational results from an ongoing phase II study [abstract]. Cancer Res. 2013;73(8 Supplement). Abstract LB-141
60
O’Brien
 
SM
Lamanna
 
N
Kipps
 
TJ
, et al. 
 
A phase II study of the selective phosphatidylinositol 3-kinase delta (PI3Kδ) inhibitor idelalisib (GS-1101) in combination with rituximab (R) in treatment-naive patients (pts) ≥65 years with chronic lymphocytic leukemia (CLL) or small lymphocytic lymphoma (SLL) [abstract]. J Clin Oncol. 2013;31(15). Abstract 7005
61
Dighiero
 
G
Maloum
 
K
Desablens
 
B
, et al. 
French Cooperative Group on Chronic Lymphocytic Leukemia
Chlorambucil in indolent chronic lymphocytic leukemia.
N Engl J Med
1998
, vol. 
338
 
21
(pg. 
1506
-
1514
)
Sign in via your Institution