Dieta y Leucemia
Dieta y Leucemia
Dieta y Leucemia
Author manuscript
Nat Rev Dis Primers. Author manuscript; available in PMC 2017 March 04.
Author Manuscript
Abstract
Chronic lymphocytic leukaemia (CLL) is a malignancy of CD5+ B cells that is characterized by
the accumulation of small, mature-appearing lymphocytes in the blood, marrow and lymphoid
tissues. Signalling via surface immunoglobulin, which constitutes the major part of the B cell
receptor, and several genetic alterations play a part in CLL pathogenesis, in addition to interactions
between CLL cells and other cell types, such as stromal cells, T cells and nurse-like cells in the
lymph nodes. The clinical progression of CLL is heterogeneous and ranges from patients who
Correspondence to T.J.K. Division of Hematology-Oncology, Department of Medicine, Moores Cancer Centre, University of
California, San Diego, 3855 Health Sciences Drive M/C 0820, La Jolla, California 92093, USA. tkipps@ucsd.edu.
Author Manuscript
Author contributions
Introduction (T.J.K.); Epidemiology (T.J.K.); Mechanisms/ pathophysiology (T.J.K., C.M.C., C.J.W., G.P. and F.K.S.); Diagnosis,
screening and prevention (K.R., W.G.W. and T.J.K.); Management (W.G.W., S.O., J.G. and T.J.K.); Quality of life (J.G. and T.J.K.);
Outlook (T.J.K.); Overview of Primer (T.J.K.).
Competing interests
J.G. has received honoraria for advisory boards from Roche, Genentech, AbbVie, Janssen, Pharmacyclics, Acerta, Gilead, TG
Therapeutics and Unum. S.O. has acted as a consultant for Amgen and Celgene, is a scientific advisory board member for CLL Global
Research Foundation and has received research support from Acerta, TG Therapeutics, Regeneron, Gilead, Pharmacyclics and
ProNAi. K.R. is a member of the medical advisory board for Celgene, Roche/Genetech, Pharmacyclics and Gilead. T.J.K. has received
honoraria for Ad boards, meetings, presentations and/or consulting from Gilead, Pharmacyclics, Celgene, Roche and AbbVie, and has
received research funding from AbbVie and Celgene. C.J.W. is a co-founder and a member of the scientific advisory board of Neon
Therapeutics. W.G.W, G.P., C.M.C. and F.K.S. declare no competing interests.
SUPPLEMENTARY INFORMATION
See online article: S1 (table) | S2 (table)
Kipps et al. Page 2
require treatment soon after diagnosis to others who do not require therapy for many years, if at
Author Manuscript
all. Several factors, including the immunoglobulin heavy-chain variable region gene (IGHV)
mutational status, genomic changes, patient age and the presence of comorbidities, should be
considered when defining the optimal management strategies, which include chemotherapy,
chemoimmunotherapy and/or drugs targeting B cell receptor signalling or inhibitors of apoptosis,
such as BCL-2. Research on the biology of CLL has profoundly enhanced our ability to identify
patients who are at higher risk for disease progression and our capacity to treat patients with drugs
that selectively target distinctive phenotypic or physiological features of CLL. How these and
other advances have shaped our current understanding and treatment of patients with CLL is the
subject of this Primer.
CLL can be divided into two main subsets, which differ in their clinical behaviour. These
subsets are distinguished by whether CLL cells express an unmutated or mutated
immunoglobulin heavy-chain variable region gene (IGHV), reflecting the stage of normal B
cell differentiation from which they originate1,2. CLL cells that express an unmutated IGHV
originate from a B cell that has not undergone differentiation in germinal centres, which are
the sites in the lymph nodes where B cells experience somatic hypermutation in their
immunoglobulin variable region genes and selection during an immune response. Patients
Author Manuscript
with CLL cells that express an unmutated IGHV typically have more-aggressive disease than
patients with CLL cells that express a mutated IGHV. CLL cells with mutated IGHV arise
from a post-germinal centre B cell that expresses immunoglobulin that has undergone
somatic hypermutation and, in some cases, also immunoglobulin isotype switching (FIG. 1),
similar to what occurs in normal B cells during an immune response to antigen. It should be
emphasized that the high level of somatic mutations that arise in IGHV in the germinal
centre are a natural part of affinity maturation of antibodies and, unlike mutations in other
genes, are not pathological. The tumours are simply reflecting the stage of maturation of the
parental B cell. In addition, some CLL cells have been described that are similar to
unmutated IGHV CLL, but originate from B cells with limited somatic mutation, such as
CLL with immunoglobulin heavy chains encoded by mutated IGHV3–21 and
immunoglobulin light chains encoded by unmutated IGLV3–21 (REFS 3,4).
Author Manuscript
The repertoire of immunoglobin molecules produced by the CLL cells of all patients is
considerably more limited than the repertoire of immunoglobulin molecules that can be
made by the B cells of any one person5,6, reflecting the biased use in CLL of certain IGHV
genes that have restricted somatic mutation and limited junctional and heavy-light chain
combinatorial diversity. In as many as one-third of patients, the CLL cells express
immunoglobulin ‘stereotypes’, which are stretches of primary structure in the variable
Nat Rev Dis Primers. Author manuscript; available in PMC 2017 March 04.
Kipps et al. Page 3
region that can also be identified in the immunoglobulins produced by the CLL cells of other
patients7. The restricted immunoglobulin repertoire in CLL is underscored by the finding
Author Manuscript
that ~1 in 75 patients have CLL cells that express immunoglobulin molecules that are
virtually identical8. The limited immunoglobulin diversity provides compelling evidence that
CLL B cells are selected based on the binding activity of their expressed surface
immunoglobulin, suggesting that B cell receptor (BCR) signalling plays a crucial part in
CLL pathogenesis.
Several large genetic studies have revealed numerous genetic alterations in CLL, including
single- nucleotide polymorphisms (SNPs), chromosomal alterations and alterations in non-
coding RNA, such as microRNA (miRNA), some of which can be used to determine
prognosis and to guide management strategies. Interactions between CLL cells and their
microenvironment, including interactions with other cell types, such as T cells, nurse-like
cells and stromal cells, can induce B cell proliferation and contribute to disease.
Author Manuscript
The distinctive cytogenesis of CLL contrasts with most other B cell malignancies, such as
follicular lymphoma, which is a germinal centre neoplasm, or myeloma (a post-germinal
centre neoplasm)9,10. However, diffuse large B cell lymphoma (DLBCL) resembles CLL in
consisting of two main subtypes: a germinal centre B-type DLBCL, which is derived from
germinal centre light zone B cells, and an activated B cell (or non-germinal centre) DLBCL,
which is derived from a later stage of germinal centre differentiation (before plasmablastic
differentiation)10. As in CLL, these two subtypes of DLBCL generally have distinctive
responses to therapy and clinical outcomes.
In this Primer, we describe the molecular pathogenesis of CLL and discuss the current
advances that are shaping our understanding and treatment of patients with this disease.
Author Manuscript
Epidemiology
CLL is estimated to account for ~19,000 of all newly detected cancers in the United States
in 2016 (REF. 11). The average incidence of CLL varies between individuals in different
geographical regions and ranges from <0.01% of individuals in eastern Asia to ~0.06% of
individuals in Europe and the United States. The risk of developing CLL is about two-times
higher for men than for women and increases with age; the median age at diagnosis ranges
from 70 to 72 years11–14.
The US National Cancer Institute Surveillance, Epidemiology, and End Results programme
has estimated the number of new cases of CLL to be 6.3 per 100,000 men and 3.3 per
100,000 women. The incidence in white populations is estimated to be 6.8 per 100,000 men
Author Manuscript
and 3.5 per 100,000 women, 4.9 per 100,000 men and 2.4 per 100,000 women in African
Americans, 2.7 per 100,000 men and 1.6 per 100,000 women in Hispanic Americans, 1.7 per
100,000 men and 1.3 per 100,000 women in Indigenous Americans, and 1.7 per 100,000
men and 0.3 per 100,000 women in people of Asian or Pacific Island descent in the United
States13.
Nat Rev Dis Primers. Author manuscript; available in PMC 2017 March 04.
Kipps et al. Page 4
Hereditary factors
Author Manuscript
Genetic factors contribute to disease susceptibility; among patients who are registered in the
CLL Research Consortium, 9% of patients have a relative with CLL. In addition, first-degree
relatives of patients with CLL have an 8.5-fold increased risk of developing this disease15,
and the concordance of CLL is higher among monozygotic twins than among dizygotic
twins16. Genome-wide association studies have identified SNPs in nearly 30 loci that are
associated with familial CLL, demonstrating that common genetic variation contributes to
heritable risk17–22 (see Supplementary information S1 (table)).
The altered expression of genes that are located in or near CLL-associated SNPs might
contribute to disease development. For example, a SNP in IRF4 is associated with low
expression of interferon regulatory factor 4; mice that are deficient in this protein can
develop CLL23, partly owing to hyperactivation of Notch signalling24. SNPs in LEF1 might
Author Manuscript
Environmental factors
The US Department of Veterans Affairs has accepted that exposure to Agent Orange is a risk
factor for CLL, which has enabled veterans with CLL to claim benefits if they were
previously exposed to Agent Orange while in military service33. In addition, evidence
suggests that exposure to insecticides might be a risk factor for CLL34. By contrast, little
evidence indicates that ionizing radiation can increase the risk of CLL35,36. Furthermore,
there is scant evidence that viral infections are risk factors, and epidemiological studies have
not found evidence that blood transfusions can transmit CLL37. No evidence suggests that
Author Manuscript
Mechanisms/pathophysiology
Genetics
Genetic alterations in CLL can include chromosomal alterations, mutations, alterations in
the expression of mi RNAs and epigenetic modifications.
Nat Rev Dis Primers. Author manuscript; available in PMC 2017 March 04.
Kipps et al. Page 5
(q13;q32) or other genetic alterations that enhance the expression of CCND1, which encodes
the cell cycle regulator cyclin D1.
The functional role of several putative driver mutations has been confirmed; for example,
silencing mutated WNT pathway genes in primary CLL cells resulted in decreased cell
viability47. Mutations in POT1, which has a role in the protection of telomeres, prevented
the binding of protection of telomeres protein 1 to telomeric DNA, resulting in numerous
chromosomal abnormalities, in addition to the development of abnormal telomeres.
Mutations in SF3B1 have been found to be associated with aberrant RNA splicing45,48,49
and an altered DNA-damage response50. SAMHD1 encodes a protein involved in the
regulation of intracellular deoxy-nucleotide pools, which are recruited to the site of DNA
damage and are probably involved in the response to DNA double-strand breaks50.
The detection of somatic mutations and their relative frequencies is variable, which possibly
Author Manuscript
reflects differences in the composition of the cohorts studied worldwide. Two seminal
studies have provided the largest sequenced collections to date51,52, in which >500 CLL
samples were characterized by whole-exome sequencing or whole- genome sequencing. The
clinical and/or biological features of the patients examined in each study were notably
distinct; one study analysed matched pretreatment samples from patients who required initial
treatment and noted mutations in SF3B1 (21% of patients), ATM (15% of patients), TP53
(7% of patients), NOTCH1 (6% of patients) or BIRC3 (4% of patients). The other study
assessed patients with earlystage CLL and patients with monoclonal B cell lymphocytosis
Nat Rev Dis Primers. Author manuscript; available in PMC 2017 March 04.
Kipps et al. Page 6
patients) and SF3B1 (8.6% of patients) as the most frequently mutated genes.
These large sample cohorts have provided the sensitivity to discover novel candidate cancer
genes that are altered in CLL. Both studies also identified somatic mutations in MGA and
PTPN11, which encode modulators of MYC, IKZF3, which encodes a key transcription
factor, and RPS15, which encodes 40S ribosomal protein S15 and is recurrently mutated in
~20% of patients who relapse after combination chemotherapy53. Other recurrent somatic
mutations include those in the 3′ untranslated region of NOTCH1 and a PAX5 enhancer,
which increases the expression of these B cell-associated transcription factors54,55. Patients
with mutations in the 3′ untranslated region of NOTCH1 have a shorter time from diagnosis
to treatment and poorer overall survival, similar to that of patients with non- synonymous
mutations, which alter the amino acid sequence of NOTCH1.
Author Manuscript
one study of CLL cells from patients treated with ibrutinib (an inhibitor of Bruton tyrosine
kinase (BTK)), revealed mutations associated with drug resistance that were distinct from
those observed in CLL cells of patients treated with standard chemotherapy58.
miRNA alterations—CLL was the first human disease that was found to be associated
with alterations in miRNA. Specifically, mir-15a and mir-16-1 (REF. 59) are deleted, altered
or downregulated in ~60% of patients with CLL and are dysfunctional in a few cases of
familial CLL26. mir-15a and mir-16-1 both target BCL2 and MCL1 (REF. 28), which
encode anti-apoptotic proteins of the BCL-2 family60; reduced expression or loss of these mi
RNAs can enhance the expression of these target genes. Attention has also focused on mi
RNAs that are dysregulated or that are differentially expressed in subgroups with distinctive
clinical and/or biological features61 (see Supplementary information S2 (table)). For
Author Manuscript
example, miR-29a/b, miR-29c, miR-34b, miR-181b and miR-3676 target the 3’ untranslated
region of TCL1A62; loss or reduced expression of all or some of these miRNAs can lead to
enhanced expression of TCL1A, which, when constitutively expressed in mature B cells,
promotes the development of CLL in transgenic mice63. By contrast, increased expression of
mir-155 is associated with enhanced BCR signalling, B cell proliferation and
lymphomagenesis64,65.
Nat Rev Dis Primers. Author manuscript; available in PMC 2017 March 04.
Kipps et al. Page 7
normal B cell from which the CLL cells were derived75. These studies revealed that the CLL
cells of different patients derive from a continuum of B cell maturation states, which are not
restricted to discrete maturation stages. Nevertheless, CLL cells that use unmutated IGHV
versus mutated IGHV generally have distinctive methylation patterns, which respectively
approximate to those of pre-germinal centre versus post-germinal centre memory B cells, as
depicted in FIG. 1. The diversity in the likely cells of origin of CLL cells highlights the
biological and phenotypic heterogeneity of this disease. These findings also suggest that
epigenetic programming that is dependent of transcription factors has a potentially important
role in the development of CLL.
(either IgA, IgD, IgE, IgG or IgM) and the signalling Igα (also known as CD79A)–Igβ
(also known as CD79B) heterodimer. CLL cells typically co-express IgD and IgM, although
at low levels compared with normal B cells; less than a few per cent of CLL cases express
class-switched isotypes, most commonly IgG. The CD79A and CD79B molecules contain
immunoreceptor tyrosine-based activation motifs, which can be phosphorylated following
the crosslinking of surface immunoglobulin, thereby triggering BCR signalling. A functional
BCR is required for the survival of mature B cells76 and is maintained in most mature B cell
malignancies, including CLL. In CLL, evidence suggests that the surface immunoglobulin of
CLL B cells is engaged by autoantigen, which leads to constitutive BCR signalling in
vivo77–79. The importance of this interaction is underscored by the clinical success of kinase
inhibitors that block BCR signalling (see Management), although effects on other receptors
might also have a role80.
Author Manuscript
Like most cancers, CLL is heterogeneous and the outcome of BCR signalling ranges from
enhanced B cell activation to B cell anergy81,82. The main pathways that lead to cell survival
and proliferation downstream of the BCR are shown in FIG. 3, along with drugs targeted
against key signalling intermediates. BCR signalling that leads to anergy is less well defined,
but seems to involve biased activation of inhibitory molecules with only partial activation of
the pathways that are typically associated with B cell activation81. One important molecule
Nat Rev Dis Primers. Author manuscript; available in PMC 2017 March 04.
Kipps et al. Page 8
that may be involved is the inositol lipid phosphatase SHIP1. SHIP1 is activated by the
Author Manuscript
Enhanced B cell activation is more commonly observed in CLL that expresses unmutated
IGHV, whereas anergy predominates in most cases of CLL that express mutated IGHV84.
Anergy is a state of cellular lethargy induced by chronic engagement of the surface antigen
receptors in the absence of adequate T cell help. Although capable of reversing their
phenotype, anergic cells are less likely to proliferate in response to BCR signalling than
more activated cells, which might, in part, account for the observation that patients with
CLL cells that express mutated IGHV generally have more indolent disease than patients
with CLL cells with unmutated IGHV85. The fate of the cell (activation versus anergy)
might be influenced by the CLL cell of origin (FIG. 1), as the cell types that can form CLL
Author Manuscript
differ in their patterns of DNA methylation73, and are likely to respond differently to
autoantigens. An unresolved question is whether anergy can be reversed in vivo, mirroring
what occurs in vitro78.
The BCR also coordinates the activity of other cell surface receptors, including integrins,
such as α4β1 integrin. BCR stimulation can result in increased adhesion of CLL cells to
α4β1 integrin substrates, for example, fibronectin and vascular cell adhesion protein 1 (REF.
86). By contrast, CXC-chemokine receptor 4 (CXCR4) is downmodulated by BCR
engagement and both can trigger ‘inside-out’ signalling, resulting in the activation of α4β1
integrin87,88. Thus, recognized antigen encountered in lymphoid tissue is likely to affect
adhesion and migration of CLL cells. Modulation of these pathways, coupled with the role
of BTK and PI3K in chemokine receptor signalling89, contribute to the increased
Author Manuscript
Cancer microenvironment
CLL cells depend on survival signals that they receive in lymphoid tissues from
neighbouring non-neoplastic cells within the so-called cancer microenvironment. CLL cells
follow chemokine gradients into lymph nodes, where they form ‘proliferation centres’ (REF.
77), as opposed to normal germinal centres. In these proliferation centres, the CLL cells
contact non- malignant stromal cells, nurselike cells (also known as lymphoma-associated
macrophages), T cells and mesenchymal-derived stromal cells (FIG. 4). Engagement with
autoantigen may occur during this transit, thereby stimulating CLL cell activation and
proliferation if sufficient T cell help is available. Only a few per cent of the CLL cells
Author Manuscript
undergo proliferation at any one time; the remainder of the cells are either unstimulated or
driven into anergy84. However, within such proliferation centres, all CLL cells are exposed
to chemokines, integrins, cytokines and survival factors (such as tumour necrosis factor
(TNF) ligand super-family member 13B (also known as BAFF) or TNF ligand superfamily
member 13 (also known as APRIL)), which activate canonical nuclear factor-κB (NF-κB)90,
before they exit to the blood. Activation of NF-κB can induce the expression of mir-155,
which enhances BCR signalling and activation by reducing the expression of INPP5D,
Nat Rev Dis Primers. Author manuscript; available in PMC 2017 March 04.
Kipps et al. Page 9
which encodes SHIP1 (REF. 65). Cytokines that are secreted by T cells, such as IL-4, can
Author Manuscript
upregulate surface IgM, which potentially facilitates the interaction of the CLL cell with
autoantigen91. In addition, the elaboration of various WNT proteins by cells in the
microenvironment can activate canonical and non-canonical WNT signalling pathways92,93.
Activation of the tyrosine-kinase-like transmembrane receptor ROR1 by WNT5A can induce
the activation of RAC1 and RHOA, and thereby enhance CLL cell proliferation and promote
migration in response to chemokines93; in part, for this reason, high-level CLL cell
expression of ROR1 is associated with accelerated disease progression94. Finally, Notch
signalling in response to Jagged, or Hedgehog signalling in response to Sonic Hedgehog or
Desert Hedgehog, can provide pro-survival stimulation for at least a subset of patients with
CLL, particularly those with trisomy 12 (REFS 95–97).
As CLL cells leave the tissue site, engagement with antigen will be transient and its effects
are likely to reverse in the blood, leading to variable increases in the expression of surface
Author Manuscript
IgM and CXCR4 (REF. 98). CLL cell expression of CXCR4 is downmodulated upon
exposure to CXC-chemokine ligand 12 (CXCL12)99, which is produced by nurse-like cells
in the microenvironment100. Consequently, CLL cells in the blood that have just exited
lymphoid tissue express low levels of CXCR4 (known as CXCR4dim cells) and higher levels
of CD5 (known as CD5bright cells) relative to the CLL cells that are poised to re-enter
lymphoid compartments101. For unexplained reasons, a high level of expression of CXCR4
by circulating CLL cells is associated with poorer prognosis in patients with CLL that use
mutated IGHV102, possibly by influencing tissue re-entry. In terms of treatment effects,
kinase inhibitors, such as ibrutinib, inhibit BCR-associated pathways, which remain
important for cancer cells that are retained in lymphoid tissues, but can also directly inhibit
integrin-mediated and chemokine- mediated pathways, thereby contributing to the increased
lymphocytosis that occurs following the initiation of kinase inhibitor therapy103.
Author Manuscript
Immune deficiency
One clinically important aspect of CLL is the development of hypogammaglobulinaemia
with consequent risk of infection. The mechanism involved is unclear, but IL-10, a known T
cell-derived immunosuppressive factor, might have a role104. For CLL, emerging evidence
suggests that the cancer cells themselves can produce IL-10 (REF. 105). Apparently, more
IL-10 is produced by CLL cells that express mutated IGHV than by CLL cells that express
unmutated IGHV. However, systemic levels of IL-10 and other suppressive factors can also
be influenced by the cumulative total-body numbers of cancer cells, which are often higher
in patients with CLL cells that express unmutated IGHV. This might account in part for the
finding of immune deficiency in patients with CLL cells that express either mutated IGHV
or unmutated IGHV. Furthermore, CLL cells express high levels of programmed cell death 1
Author Manuscript
ligand 1 (PD-L1) and PD-L2, which suppress the effector responses of T cells that express
programmed cell death protein 1 (PD-1), leading to an ‘exhausted’ T cell phenotype and
impaired cellular immune function106.
Nat Rev Dis Primers. Author manuscript; available in PMC 2017 March 04.
Kipps et al. Page 10
Diagnostic work-up
Most often, patients with CLL are asymptomatic at the time of diagnosis and become aware
of the disease following the detection of lymphocytosis in a routine blood count. However,
CLL can have a range of clinical presentations; some patients feel well and are fully active,
but a minority have disease-related symptoms. The usual symptoms of CLL include fatigue,
involuntary weight loss, excessive night sweats, abdominal fullness with early satiety and
increased frequency of infections, which might be associated with
hypogammaglobulinaemia. Some patients can present with symptoms of an autoimmune
cytopenia (for example, autoimmune haemolytic anaemia or immune thrombocytopenic
purpura). Patients can also have or develop enlarged lymph nodes, hepatomegaly and
splenomegaly, which are palpable on physical examination. Enlarged lymph nodes can be
Author Manuscript
easily palpable at three sites: the cervical, axillary and inguino-femoral regions.
Laboratory features—Laboratory assessment for CLL includes a full blood cell count
and flow cytometry. The most consistent laboratory abnormality observed is an increase in
the absolute number of blood lymphocytes above the normal adult upper limit of ~3,500
cells per µl, detected by a blood count. Most patients present with ≥10,000 cells per µl, but
some might have fewer numbers of blood lymphocytes upon relapse after therapy. The initial
diagnosis requires detection of ≥5,000 cells per µl of clonal CLL B cells107, which typically
express low levels of surface immunoglobulin with either κ-immunoglobulin or λ-
immunoglobulin light chains.
1). CLL B cells typically express CD5, CD19 and CD23 (also known as low-affinity
immunoglobulin-ε Fc receptor), and have low levels of CD20, but lack expression of CD10
and stain poorly, if at all, with the FMC7 monoclonal antibody, which recognizes an epitope
of CD20 (REF. 108). CLL cells also express CD200 (also known as OX-2 membrane
glycoprotein), which can help to distinguish CLL from mantle cell lymphoma109. In
addition, the CLL cells of >95% of patients express the onco-embryonic surface antigen
ROR1 (REFS 94,110).
Morphologically, CLL cells are small mature-appearing lymphocytes with dense chromatin,
a nucleus that virtually fills the cell with only a rim of visible cytoplasm and no (or
occasionally small) nucleoli (FIG. 5a). In CLL, the presence of smudged cells on the blood
smear is common and represents lymphocytes that were crushed in the process of making
Author Manuscript
the slide (FIG. 5b). CLL cells also can appear as prolymphocytes, which are larger than
typical CLL cells, have less-condensed nuclei and a single prominent nucleolus (FIG. 5c).
However, if >55% of cells on the blood smear are prolymphocytes, a diagnosis of
prolymphocytic leukaemia should be considered111.
No abnormalities are considered specific for CLL in the blood chemistry panel. Quantitative
levels of serum immunoglobulins (for example, IgA, IgG and IgM) are usually normal at
diagnosis, but generally decline with disease progression. A direct Coombs test (which is
Nat Rev Dis Primers. Author manuscript; available in PMC 2017 March 04.
Kipps et al. Page 11
used to detect erythrocytes that are coated with anti-red blood cell autoantibodies) might be
Author Manuscript
Although not required for establishing a diagnosis of CLL, a marrow biopsy is often
performed; this usually shows hypercellularity owing to an increased percentage of mature-
appearing lymphocytes. Four patterns of lymphocytic infiltration in the marrow have been
described: nodular, interstitial, mixed (nodular and interstitial) or diffuse; the diffuse pattern
is typically associated with advanced disease112 (FIG. 6). In addition, the marrow usually
shows reduced numbers of myeloid and erythroid cells, which otherwise have normal
maturation.
A lymph node biopsy might be performed in a patient with an enlarged lymph node as part
Author Manuscript
of a diagnostic evaluation for suspected lymphoma. Excised lymph nodes typically have a
diffuse infiltration of well-differentiated small lymphocytes, often obliterating the normal
nodal architecture, and scattered, vaguely nodular, pale haematoxylin and eosin-staining
areas, appearing as pseudo-follicles (FIG. 7a), which are enriched with prolymphocytes and
paraimmunoblasts (FIG. 7b); these areas comprise the proliferation centres113. The
pseudofollicles or proliferation centres are hallmark features in the lymph nodes of patients
with CLL or small lymphocytic lymphoma, as they are not observed in other types of
lymphomas.
Staging
Two clinical staging systems are widely used to divide patients with CLL into three broad
prognostic groups114,115. The Rai staging system (TABLE 1) is more commonly used in the
Author Manuscript
United States, whereas the Binet classification (TABLE 2) is more commonly used in
Europe. The staging systems each recognize the importance of marrow function and define
late-stage, or high-risk, disease by the presence of pronounced anaemia or
thrombocytopenia.
Prognostic factors that can help to identify patients who require therapy relatively soon after
diagnosis include certain clinical features and genetic, molecular and biochemical
characteristics of the CLL cell. Multivariable models, prognostic indexes116–118 and
nomograms119 have been developed to consolidate such prognostic factors so that they can
more robustly predict clinical outcome. Commonly used parameters that are associated with
poorer outcome are male sex, ≥65 years of age, poor performance status due to medical
comorbidities, certain CLL cell characteristics, such as the expression of unmutated
IGHV1,2, ZAP70 (REFS 120,121), CD49d (also known as integrin α4)122 or CD38 (REF.
Nat Rev Dis Primers. Author manuscript; available in PMC 2017 March 04.
Kipps et al. Page 12
2), the presence of del(17p)38 or del(11q)123, high serum levels of β2-microglobulin (>3.5
mg per l)124, complex karyotype (that is, the presence of three or more chromosomal
Author Manuscript
Currently, the most reliable prognostic models are those developed for treatment-free
survival, as evolving treatments have yet to change the indications for therapy. Predictive
models to define overall survival with a given type of therapy are challenged by the
chronicity of CLL and the fact that patients often receive serial treatments, each of which
can affect outcome; moreover, death might be due to an indirect or unrelated cause.
Furthermore, treatment options are changing, with newly identified, highly effective agents
that are clearly prolonging survival and have activity among patients who would have been
Author Manuscript
considered high risk when the only option was conventional chemotherapy.
Management
Generally, indications to initiate therapy include pronounced disease-related anaemia or
thrombocytopenia (patients with Rai stage III or stage IV disease, or Binet stage C disease),
symptomatic lymphadenopathy and/or symptoms that are associated with active disease,
such as night sweats, fatigue, unintentional weight loss or fever without evidence of
infection107. However, when basing a treatment decision on constitutional symptoms alone,
the physician should consider other medical conditions, such as hypothyroidism,
hyperthyroidism, hypoglycaemia, chronic inflammation, uncommon opportunistic infections
or sleep disorders, including sleep apnoea.
Author Manuscript
No established absolute lymphocyte count or lymph node size alone should form the basis
for the initiation of therapy. Instead, patients who are asymptomatic with early-stage or
intermediate-stage disease (such as Rai stage I or stage II, or Binet stage A or stage B) are
not recommended for therapy unless they have symptomatic disease or evidence for disease
progression. Evidence for disease progression can include a lymphocyte doubling time of <1
year, progressive palpable lymphadenopathy and/or progressive palpable splenomegaly in
serial examinations. In the absence of indications for treatment, patients are examined for
palpable lymphadenopathy and splenomegaly and have complete blood counts at 3–12-
month intervals, the frequency of which depends on the presence of signs of disease
progression. Clinical or laboratory features of anaemia or thrombocytopenia should prompt
evaluation for autoimmune haemolytic anaemia or immune thrombocytopenic purpura,
respectively; such autoimmune cytopenias might require treatment that is independent of the
Author Manuscript
consideration for therapy directed against the under lying CLL. Finally, patients should be
cautioned to seek prompt medical attention for signs or symptoms of infection; because of
the acquired immune deficiency associated with CLL, the threshold for considering the use
of antimicrobial therapy should be low. Nonetheless, development of frequent or serious
infections is not an indication for CLL-directed therapy.
Nat Rev Dis Primers. Author manuscript; available in PMC 2017 March 04.
Kipps et al. Page 13
For patients who need treatment, the presence of del(17p) or mutated TP53 are the most
Author Manuscript
important features that are currently directing the choice of therapy (FIG. 8). Next, advanced
age of >65 years, the presence of medical comorbidities and the objectives of treatment have
substantial bearing on the choice of therapy. Increasingly, IGHV mutational status is
considered as a parameter when determining the type of therapy; for example,
chemotherapy-based regimens are reserved for patients with CLL and mutated IGHV.
Conversely, the specific Rai or Binet stage of the patient who requires treatment does not
necessarily influence the choice of therapy.
Systemic treatments
The treatment of patients with CLL can include chemotherapy, a combination of
chemotherapy and immunotherapy, or drugs that target the signalling pathways that promote
the growth and/or survival of CLL cells (for example, BCR signalling and BCL-2)128,129.
Author Manuscript
Chemotherapy—Chemotherapy has been the mainstay of therapy for the past 50 years.
Purine analogues (most commonly fludarabine, but also pentostatin or cladribine) and
alkylating agents (including chlorambucil, cyclophosphamide or bendamustine) are used in
the treatment of CLL130–132. Chemotherapy-based regimens can cause myelosuppression, an
increased risk of infections and, in a small subset of patients, post-therapy myelodysplasia or
secondary cancers, such as acute myeloid leukaemia (see Secondary cancers).
than patients who were treated with fludarabine and cyclophosphamide133. In a separate
study (the CLL11 trial), patients >65 years of age with medical comorbidities who were
treated with chlorambucil and either obinutuzumab or rituximab had improved response
rates and longer median PFS than patients who were treated with chlorambucil alone134.
However, the median PFS was significantly longer for patients who received obinutuzumab
(26.7 months) than in those who received rituximab (11.1 months). In a third phase III trial,
median PFS significantly improved from 13.1 months for patients treated with chlorambucil
to only 22.4 months for patients treated with chlorambucil and ofatumumab135. As a
consequence of these three trials, the US FDA approved the use of rituximab, obinutuzumab
or ofatumumab in combination with chemotherapy for the first-line treatment of patients
with CLL. The FDA also approved the use of ofatumumab as a single agent for the treatment
of patients with relapsed or refractory disease based on data from a phase II study136.
Author Manuscript
Bendamustine is commonly used with rituximab and has good response rates in treatment-
naive patients without del(17p)137, although no randomized trials comparing bendamustine
and rituximab versus bendamustine alone have been conducted. Bendamustine has also been
used in combination with obinutuzumab, which showed highly encouraging results138 and is
being evaluated in larger clinical trials.
Nat Rev Dis Primers. Author manuscript; available in PMC 2017 March 04.
Kipps et al. Page 14
In a randomized trial, the rates of complete response and complete response without
Author Manuscript
evidence for minimal residual disease (MRD) were higher in patients treated with
fludarabine, cyclophosphamide and rituximab than in those treated with bendamustine and
rituximab, and the median PFS was ~1 year longer139. However, patients in the
bendamustine and rituximab treatment subgroup were older and had a higher proportion of
patients who had CLL cells expressing unmutated IGHV, making this cohort at higher risk
for a poorer outcome than the cohort of patients treated with fludarabine, cyclophosphamide
and rituximab. It also should be noted that patients treated with fludarabine,
cyclophosphamide and rituximab had higher rates of neutropenia and infections than
patients treated with bendamustine and rituximab. Because of this, many physicians
currently provide patients with growth factors (for example, filgrastim or pegfilgrastim) and
prophylactic antimicrobial therapy when they are treated with the fludarabine,
cyclophosphamide and rituximab regimen, but such measures were not recommended for
patients treated in this trial139. In any case, there has not been significant difference observed
Author Manuscript
in overall survival between the two treatment arms, but events are limited.
Some patients can experience a prolonged PFS following treatment with fludarabine,
cyclophosphamide and rituximab, particularly those with CLL with mutated IGHV that lack
del(17p) or del(11q), which are associated with chemotherapy resistance or relatively short
PFS, respectively. Long-term follow-up data on patient outcomes following therapy with this
regimen indicate that patients with mutated IGHV might achieve a long-term survival
benefit (and possible ‘cure’) with chemoimmunotherapy140–142.
Inhibitors of BCR signalling—Three main classes of drugs that each can inhibit BCR
signalling have been evaluated in patients with CLL: BTK inhibitors, PI3K inhibitors and
spleen tyrosine kinase (SYK) inhibitors86,143. CLL cells with unmutated IGHV seem to be
Author Manuscript
more sensitive to inhibitors of BCR signalling than CLL cells with mutated IGHV144, but
whether inhibitors, such as ibrutinib, are more effective in patients with CLL and unmutated
IGHV, remains to be validated in clinical trials.
Ibrutinib has been approved in the United States and Europe for use as initial therapy, as
well as in patients with relapsed disease, which followed results from a randomized trial that
showed a significantly higher response rate to therapy with ibrutinib than with
ofatumumab145. In addition, with continuous therapy, patients treated with ibrutinib had a
significantly longer median PFS and overall survival than patients treated for 8 months with
ofatumumab. Approval of ibrutinib as initial therapy was based on the results of a
randomized trial that showed a significant improvement in median PFS and overall survival
in patients ≥65 years of age without del(17p) who were treated indefinitely with ibrutinib
Author Manuscript
Nat Rev Dis Primers. Author manuscript; available in PMC 2017 March 04.
Kipps et al. Page 15
arthralgia, myalgia, increased blood pressure and atrial fibrillation. Clinical trials are
currently evaluating second-generation BTK inhibitors (for example, acalabrutinib148, ONO/
GS-4059 (REF. 149) or BGB-3111) to determine whether any one of these drugs has a
superior therapeutic index than that of ibrutinib150.
PI3K inhibitors include idelalisib, duvelisib (also known as IPI-145), TGR-1022 and
ACP-319 (also known as AMG-319)151; the latter three drugs are being evaluated in clinical
trials, whereas idelalisib was approved in the United States and Europe for the treatment of
patients with relapsed CLL; this approval was based on the outcome of a clinical trial that
showed that patients treated with rituximab and idelalisib had significantly higher response
rates and a significantly longer median PFS and overall survival than patients treated with
rituximab and placebo152. As with ibrutinib, patients who initiate therapy with idelalisib can
experience a rapid reduction in lymphadenopathy that is associated with lymphocytosis.
Author Manuscript
Adverse effects of idelalisib include transaminitis (usually in the first few months of
therapy), pneumonitis and colitis; the latter usually occurs >6 months after the initiation of
therapy with this drug and is often severe enough to require cessation of therapy153.
Transaminitis seemed to be more severe in patients who received idelalisib as their initial
therapy for CLL than in patients with relapsed disease153, suggesting that transaminitis is
not directly caused by idelalisib. This is also suggested by the observations that mild
increases in the levels of serum transaminase can subside over time with continued drug
administration; furthermore, patients who have had idelalisib withheld because of
transaminitis can be restarted on this drug without experiencing apparent hepatic toxicity.
The decision to halt therapy or to re-administer the drug following resolution of
Author Manuscript
transaminitis should consider the severity and duration of hepatic function test
abnormalities, which often do not recur upon re-institution of idelalisib therapy154.
In 2016, the FDA recommended the closure of clinical trials investigating idelalisib and
rituximab combination therapy for first-line treatment of patients with CLL, owing to a
higher number of infections and deaths in the experimental arm. As such, patients
undergoing therapy with idelalisib and rituximab should be considered for concomitant
treatment with prophylactic low-dose acyclovir to protect against reactivation of varicella
zoster virus, which causes chicken pox and shingles. Patients also should be treated with
prophylactic antibiotics to mitigate the risk for opportunistic infection, such as that caused
by Pneumocystis jiroveci. Finally, as with any patient receiving therapy with anti-CD20
monoclonal antibodies, patients should be screened for active infection with hepatitis B
virus before the initiation of therapy155, and periodically monitored for reactivation of
Author Manuscript
Phase I/II clinical trials of fostamatinib, an oral SYK inhibitor, caused reduction in
lymphadenopathy with concomitant lymphocytosis, an improvement in disease-related
cytopenias and relief of disease-related symptoms in most of the treated patients with
CLL156. However, dose-limiting toxicities of fostamatinib treatment include neutropenia,
Nat Rev Dis Primers. Author manuscript; available in PMC 2017 March 04.
Kipps et al. Page 16
thrombocytopenia and diarrhoea. Other inhibitors of SYK, such as entospletinib, are being
Author Manuscript
dose escalation.
Assessment of response
Historically, a favourable response to therapy has been defined as a partial remission or
complete remission. Partial remission requires a 50% reduction in tumour bulk (for example,
lymphadenopathy and splenomegaly), a 50% reduction in lymphocytosis, and platelet counts
of >100,000 cells per µl (or 50% improvement over baseline) or a haemoglobin level of >11
g per dl (or 50% improvement over baseline) without requiring transfusions or exogenous
growth factors107. Complete remission requires the normalization of blood counts,
resolution in lymphadenopathy and splenomegaly, and normal marrow function. The use of
CT to assess response in CLL is becoming more common, particularly in clinical trials.
However, the benefit of using repeated CT scans to monitor disease is uncertain, and seems
Author Manuscript
unlikely to change patient outcome. Because of the distinct pattern of response observed
with BCR inhibitors, a new response category, namely, partial response with lymphocytosis,
has been described. Partial response with lymphocytosis is defined as a >50% reduction in
lymphadenopathy and splenomegaly, with persistent lymphocytosis; often the blood
lymphocyte counts are equal to or greater than those observed prior to therapy.
Nat Rev Dis Primers. Author manuscript; available in PMC 2017 March 04.
Kipps et al. Page 17
In clinical trials, it is becoming more common to evaluate for MRD with ≥0.01% of CLL
Author Manuscript
cells among the total population of mononuclear cells in the blood or marrow. MRD can be
measured by flow cytometry or PCR with next-generation sequencing of the clonal
immunoglobulin variable region gene rearrangements163. In most clinical trials for patients
with CLL, particularly those conducted in Europe, evaluation of MRD has been performed
by flow cytometry of mononuclear cells from the marrow aspirate (the preferred method) or
from the peripheral blood. In the 6 months following anti-CD20 monoclonal antibody
treatment, the assessment of MRD is more sensitive on the mononuclear cells of the marrow
aspirate than on cells that are isolated from the blood, which will often lack detectable CLL
even when they are readily found in the marrow. Beyond a complete response, the best
predictor of long-term PFS and overall survival is the achievement of a complete remission
without evidence for MRD.
Relapsed disease
Author Manuscript
The treatment landscape for relapsed and refractory CLL will be changing owing to the first-
line approval of ibrutinib. Currently, most patients with relapsed or refractory disease receive
chemoimmunotherapy. Standard salvage regimens include BCR inhibitors or BCL-2
inhibitors, particularly for patients with CLL and del(17p). For patients who received first-
line BTK inhibitor therapy, salvage options include chemo immunotherapy (fludarabine,
cyclophosphamide and rituximab (or bendamustine with an anti-CD20 monoclonal
antibody), PI3K inhibitor and an anti-CD20 monoclonal antibody152, high-dose
methylprednisolone and an anti-CD20 monoclonal antibody164, or lenalidomide alone or
with an anti-CD20 monoclonal antibody, although lenalidomide has not been approved for
the treatment of patients with CLL by the FDA165,166. Treatment choice depends on
individual patient characteristics and the intent of treatment. Scant data are available
Author Manuscript
regarding the activity of small-molecule inhibitors in patients who are refractory to another
small-molecule inhibitor; better efficacy is expected for patients who discontinued use of
another small-molecule inhibitor due to intolerance167. Ibrutinib resistance is an adverse
predictor of clinical outcome, particularly for patients who were previously exposed to
chemoimmunotherapy. If a previously treated patient develops del(17p), or mutated TP53,
treatment options include ibrutinib, venetoclax, or idelalisib and an anti-CD20 monoclonal
antibody. The patient could also participate in a clinical trial. The preference for non-
chemotherapy-based treatment should be driven by prior exposure to a small-molecule
inhibitor and a review of the safety profile of the drug.
currently not considered the standard of care, but might be useful in patients with medical
comorbidities that limit other treatment options.
Nat Rev Dis Primers. Author manuscript; available in PMC 2017 March 04.
Kipps et al. Page 18
Quality of life
Author Manuscript
Comorbidities
As CLL is a disease of the elderly population, assessing the effect of CLL on the patient’s
quality of life (QOL) and the coexisting comorbidities that occur in this patient population is
important169. Awareness regarding the importance of a patient’s QOL, not only during and
after treatment but also during the watch-and-wait period, is increasing.
Until recently, few clinical trials included elderly or frail patients, who account for most
patients with CLL. As such, the recommendations for therapy were largely based on results
from clinical trials that were conducted with younger patients who could better tolerate
combination drug therapies. Trials have moved away from using eligibility criteria based on
age or creatinine clearance, to using more objective measures of fitness, such as the
cumulative illness rating score, which can stratify patients for appropriate first-line or
Author Manuscript
subsequent therapy. This has led to an increased number of published clinical trials targeting
patients who would not be deemed fit for aggressive chemoimmunotherapy approaches.
Importantly, this has also demonstrated that ‘unfit’ patients with CLL can be recruited to
clinical trials in a timely manner.
quantitative and functional defects170. The risk of infections increases with worsening
hypogammaglobulinaemia. The degree of immune impairment worsens with disease
progression and can be exacerbated by the immunosuppressive effects of purine analogue
chemotherapy, anti-CD20 monoclonal antibodies or drugs that inhibit kinases involved in
immune receptor signalling. Consequently, infectious complications represent a frequent
cause of morbidity and mortality in patients with CLL.
Infections are typically bacterial and frequently involve the respiratory tract. Intravenous
immunoglobulin replacement therapy can mitigate the risk of infection, particularly in
patients with hypogammaglobulinaemia who have frequent infections or a severe life-
threatening infection171. Immunoglobulin formulations that are administered subcutaneously
seem to be as effective and might be less costly172. Unfortunately, randomized studies
Author Manuscript
Nat Rev Dis Primers. Author manuscript; available in PMC 2017 March 04.
Kipps et al. Page 19
might worsen immune function173. Finally, because of their suppressed immune function,
Author Manuscript
patients should avoid having live vaccines, such as those used to vaccinate against shingles.
No systematic controlled trials of treatment for autoimmune cytopenias in patients with CLL
Author Manuscript
have been conducted. Corticosteroids remain the mainstay of initial treatment, but
mycophenolate and thrombopoietin-like agents might be helpful for patients with immune
thrombocytopenic purpura175,176. Second-line treatments include cyclosporine or rituximab.
Splenectomy can be helpful in patients with severe or recurrent immune cytopenias who are
good-risk surgical candidates, but risks further impairment of immune function. Refractory
autoimmune haemolytic anaemia or immune thrombocytopenic purpura might require
treatment of the underlying CLL, preferably with therapy that does not substantially impair
compensatory haematopoiesis.
for melanoma, sarcomas, and lung, renal and prostate cancers177. The immune deficiencies
that are associated with CLL might contribute to this increased risk, but the malignancies
observed do not mirror those in patients with other immune-deficiency diseases. Exceptions
to this observation are Merkel cell carcinoma178, which is associated with Merkel cell
polyomavirus infection, and Bowen disease, which is an aggressive form of squamous cell
carcinoma associated with human papillomavirus infection179. Although initial studies had
suggested that the risk of secondary cancers was increased following chemotherapy,
Nat Rev Dis Primers. Author manuscript; available in PMC 2017 March 04.
Kipps et al. Page 20
subsequent studies have suggested that the risk is similar in untreated patients who continue
on watch and wait180.
Author Manuscript
Nat Rev Dis Primers. Author manuscript; available in PMC 2017 March 04.
Kipps et al. Page 21
normal haematopoietic cells to genotoxic stress, will decrease the incidence of this serious
Author Manuscript
complication.
Outlook
The outlook for patients with CLL has improved substantially over the past several years.
Through research on the immune biology and genetics of CLL, patients can be stratified into
subgroups with distinctive clinical features, which has improved our capacity to assess
prognosis or govern therapy. However, an understanding of the mechanisms that contribute
to immune dysfunction or how it contributes to autoimmune disease, such as autoimmune
haemolytic anaemia, therapy resistance or therapy-related complications is unknown.
Whether tyrosine kinase inhibitors can affect clonal evolution, induce and/or select for drug
resistance, or can achieve complete responses if used earlier in the course of the disease is
also unknown.
Author Manuscript
Future treatments
Several therapies are currently under preclinical and clinical investigation for the treatment
of patients with CLL, including new drugs and treatment modalities that can modulate the
immune system, and cell transplantation.
Thalidomide has little activity in patients with CLL as a monotherapy, but has shown
efficacy when combined with other drugs, such as anti-CD20 monoclonal antibodies190.
Author Manuscript
Conversely, lenalidomide monotherapy has an overall response rate of 60% (15% complete
response rate) as a first-line therapy and 40% (8% complete response rate) as a salvage
treatment165,189. However, trials assessing the use of lenalidomide as a monotherapy or
combination therapy have yielded mixed results. One phase II study reported that
lenalidomide was well tolerated as initial therapy in patients >65 years of age, with an
overall response rate of 65% and a complete response rate of 10%189. However, a
multicentre phase III trial for the same patient population had to be terminated owing to an
increased number of deaths in patients receiving lenalidomide compared with those
Nat Rev Dis Primers. Author manuscript; available in PMC 2017 March 04.
Kipps et al. Page 22
with rituximab was well tolerated in a multi-institutional phase II study and yielded higher
response rates; for patients <65 years of age, the overall response rate was 95%, with a 20%
complete response rate, and for patients ≥65 years of age, the overall response rate was 78%,
with an 11% complete response rate166. The PFS after completion of therapy was ~20
months for both groups. Therapy with an anti-CD20 monoclonal antibody 9 days before initi
ation of lenalidomide therapy also seems to mitig ate the risk for tumour flare
reaction192,193. The use of lenalidomide after chemoimmunotherapy has been evaluated in a
phase III maintenance trial (Continuum Trial; ClinicalTrials.gov identifier: NCT00774345)
and results are forthcoming. Ongoing trials are also examining the activity in CLL of a novel
lenalidomide analogue, CC-122 (REF. 194).
with high-risk features such as del(17p). In two clinical studies with patients who lacked
serious medical comorbidities and had a median age of 53 or 58 years, the PFS at 3–5 years
was 40–50% and overall survival was 50–70%, but the non-relapse mortality at 3–5 years
was 25–40%195,196. Research efforts are ongoing to develop better-tolerated cell-based
therapy with a similar curative potential that can be used without the immunosuppression
and associated long-term morbidity and mortality of allogeneic stem cell transplantation.
desirable option for most patients. Ongoing discussion exists around who are the appropriate
patients for allogeneic stem cell transplantation.
T cell therapy with chimeric antigen receptors—T cells can be modified ex vivo to
express new surface receptors, known as chimeric antigen receptors (CARs), which have
been engineered to target cancer cells, expanded in vitro and then reintroduced into the
patient as a treatment for CLL (BOX 3).
Both normal and malignant B cells (including CLL cells) express surface CD19, which has
been targeted with CAR technology. CD19-targeted CAR T cells have yielded long-term
PFS and relapse-free survival durations in patients with CLL; in 14 patients with relapsed or
refractory CLL, four patients achieved a complete response and four patients achieved a
partial response197. None of the complete responders had MRD and none relapsed (a median
Author Manuscript
follow-up of 19 months). However, the efficacy of CAR T cell therapy in patients with CLL
has been modest compared with that in patients with acute lymphoblastic leukaemia; this
might be owing to qualitative defects in the T cells of patients with CLL, who are generally
older than patients with acute lymphoblastic leukaemia and already have immune
dysfunction that reflects disease-associated anergy (see BCR and B cell signalling). Ibrutinib
might partially correct some of these defects198. Larger phase II trials assessing the use of
Nat Rev Dis Primers. Author manuscript; available in PMC 2017 March 04.
Kipps et al. Page 23
CAR T cell therapy for CLL are in development, including the use of CAR T cells that can
Author Manuscript
The major adverse effect of CAR T cell therapy is a cytokine release syndrome, which
occurs as a result of CAR T cell activation, cytokine production and T cell expansion
following target antigen encounter. Cytokine release syndrome is characterized by fever,
hypotension and capillary leakage, but neurological toxicity, which can manifest as
confusion and seizures, has also been observed in some of the treated patients. Cytokine
release syndrome is associated with high cytokine levels, particularly IL-6, and can be
managed with an IL-6-binding factor, such as tocilizumab, supportive measures, and
glucocorticoids for severe cases. This syndrome seems to be proportionate to the antigen-
bearing tumour burden, potentially making CAR T cell therapy more amenable to treatment
of patients with MRD.
Author Manuscript
The immune checkpoint receptor PD-1, and its ligands PD-L1 and PD-L2, is the most
important cognate receptor involved in the suppression of cellular immune activation. CLL
cells express high levels of PD-L1 and PD-L2 and can suppress the responses of PD-1-
expressing effector T cells106, leading to an exhausted (that is, no longer functional) T cell
phenotype. T cell exhaustion in CLL is also mediated in part by lymphocyte activation gene
Author Manuscript
3 protein and T cell immunoglobulin mucin receptor 3 (TIM3; also known as HAVCR2).
TIM3 negatively regulates the function of type 1 helper T cells and type 1 CD8+ T cells, by
triggering cell death upon ligand binding. Other receptors on CLL cells have demonstrated
negative immune feedback, including CD276, CD200 and TNF receptor superfamily
member 14.
A partial list of immune checkpoint inhibitors that are being evaluated in the therapy of
patients with CLL or other cancers include monoclonal antibodies against PD-1, cytotoxic T
lymphocyte protein 4, B lymphocyte and T lymphocyte attenuator and its ligand TNF
superfamily member 14, the adenosine A2A receptor, indoleamine 2,3-dioxygenase, the V-
type immunoglobulin domain-containing suppressor of T cell activation, lymphocyte
activation gene 3 protein and TIM3.
Author Manuscript
Preclinical studies in mouse models have demonstrated that checkpoint inhibitors can
reactivate immune effector cells to have anti-leukaemia activity202. However, ongoing phase
I/II trials of immune checkpoint inhibitors in patients with relapsed CLL have yet to show
much clinical activity, possibly reflecting the highly immune suppressive nature of CLL
cells and/or the ‘exhausted’ phenotype of T cells in patients with this disease.
Nat Rev Dis Primers. Author manuscript; available in PMC 2017 March 04.
Kipps et al. Page 24
physiological features of CLL with targeted therapeutic agents, which have a higher
therapeutic index than standard chemotherapy. Through the use of combination therapy,
which targets different B cell survival signalling pathways and/or achieves better eradication
of CLL cells, we might be able to define curative treatments for most patients with this
disease.
CLL cells and accessory cells in the microenvironment might enhance CLL cell expression
of anti-apoptotic proteins other than BCL-2, such as MCL1, thereby contributing to therapy
resistance. As such, the therapeutic use of a selective BCL-2 antagonist, such as venetoclax,
might be more effective when used in combination with BCL-2 inhibitors161,162, which also
interfere with the homing of CLL cells to the microenvironment. Conceivably, combination
target therapy with agents that have synergistic activity will provide highly effective and
potentially curative treatment of patients with CLL.
Supplementary Material
Refer to Web version on PubMed Central for supplementary material.
Author Manuscript
Acknowledgments
The authors thank H.-Y. Wang and E. Broome, Department of Pathology, University of California San Diego, for
their contributions to Figures 5–7. The authors also thank A. Greaves, University of California San Diego, for help
in figure development. This Primer is supported by the US NIH PO1-CA81534 for the CLL Research Consortium;
members include K.R., W.G.W., T.J.K., C.M.C., C.J.W. and J.G. The authors also acknowledge support to F.K.S.
and G.P. from Bloodwise, Kay Kendall Leukaemia Fund, Cancer Research UK, the Southampton Experimental
Cancer Medicine Centre, and the Southampton Cancer Research UK Centre (F.K.S. and G.P.). The authors also
acknowledge support from the Leukemia & Lymphoma Society (LLS) Specialized Center of Research Program
(SCOR) grant (T.J.K.), 1R01HL116452, 1R0 1CA182461, CLL Global Reseach Foundation (W.G.W.), the Blood
Cancer Research Fund (T.J.K.), and U10CA180861.
References
1. 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; 94:1848–
Author Manuscript
Nat Rev Dis Primers. Author manuscript; available in PMC 2017 March 04.
Kipps et al. Page 25
4. Ghia EM, et al. Use of IGHV3–21 in chronic lymphocytic leukemia is associated with high-risk
disease and reflects antigen-driven, post-germinal center leukemogenic selection. Blood. 2008;
Author Manuscript
of the normal B-cell hierarchy. Leuk. Lymphoma. 2014; 55:1251–1260. [PubMed: 23998255]
10. Basso K, Dalla-Favera R. Germinal centres and B cell lymphomagenesis. Nat. Rev. Immunol.
2015; 15:172–184. [PubMed: 25712152]
11. Siegel R, et al. Cancer treatment and survivorship statistics, 2012. CA Cancer J. Clin. 2012;
62:220–241. [PubMed: 22700443]
12. Nabhan C, et al. The impact of race, ethnicity, age and sex on clinical outcome in chronic
lymphocytic leukemia: a comprehensive Surveillance, Epidemiology, and End Results analysis in
the modern era. Leuk. Lymphoma. 2014; 55:2778–2784. [PubMed: 24588735]
13. Li Y, Wang Y, Wang Z, Yi D, Ma S. Racial differences in three major NHL subtypes: descriptive
epidemiology. Cancer Epidemiol. 2015; 39:8–13. [PubMed: 25560974]
14. Pulte D, Redaniel MT, Bird J, Jeffreys M. Survival for patients with chronic leukemias in the US
and Britain: age-related disparities and changes in the early 21st century. Eur. J. Haematol. 2015;
94:540–545. [PubMed: 25315799]
15. Cerhan JR, Slager SL. Familial predisposition and genetic risk factors for lymphoma. Blood. 2015;
Author Manuscript
Nat Rev Dis Primers. Author manuscript; available in PMC 2017 March 04.
Kipps et al. Page 26
26. Calin GA, et al. A microRNA signature associated with prognosis and progression in chronic
lymphocytic leukemia. N. Engl. J. Med. 2005; 353:1793–1801. [PubMed: 16251535] This seminal
Author Manuscript
study indicates that differences in the expression of certain mi RNAs are associated with
differences in clinical outcome, providing evidence that miRNA can influence disease progression
in patients with CLL.
27. Veronese A, et al. Allele-specific loss and transcription of the miR-15a/16-1 cluster in chronic
lymphocytic leukemia. Leukemia. 2015; 29:86–95. [PubMed: 24732594]
28. Cimmino A, et al. miR-15 and miR-16 induce apoptosis by targeting BCL2. Proc. Natl Acad. Sci.
USA. 2005; 102:13944–13949. [PubMed: 16166262]
29. Chen L, et al. ZAP-70 directly enhances IgM signaling in chronic lymphocytic leukemia. Blood.
2005; 105:2036–2041. [PubMed: 15514014]
30. Raveche ES, et al. Abnormal microRNA-16 locus with synteny to human 13q14 linked to CLL in
NZB mice. Blood. 2007; 109:5079–5086. [PubMed: 17351108]
31. Ojha J, et al. Genetic variation associated with longer telomere length increases risk of chronic
lymphocytic leukemia. Cancer Epidemiol. Biomarkers Prev. 2016; 25:1043–1049. [PubMed:
27197291]
Author Manuscript
32. Medves S, et al. A high rate of telomeric sister chromatid exchange occurs in chronic lymphocytic
leukaemia B-cells. Br. J. Haematol. 2016; 174:57–70. [PubMed: 26970083]
33. Baumann Kreuziger LM, Tarchand G, Morrison VA. The impact of Agent Orange exposure on
presentation and prognosis of patients with chronic lymphocytic leukemia. Leuk. Lymphoma.
2014; 55:63–66. [PubMed: 23573826]
34. Schinasi LH, et al. Insecticide exposure and farm history in relation to risk of lymphomas and
leukemias in the Women’s Health Initiative observational study cohort. Ann. Epidemiol. 2015;
25:803–810. [PubMed: 26365305]
35. Hsu WL, et al. The incidence of leukemia, lymphoma and multiple myeloma among atomic bomb
survivors: 1950–2001. Radiat. Res. 2013; 179:361–382. [PubMed: 23398354]
36. Radivoyevitch T, Sachs RK, Gale RP, Smith MR, Hill BT. Ionizing radiation exposures in
treatments of solid neoplasms are not associated with subsequent increased risks of chronic
lymphocytic leukemia. Leuk. Res. 2016; 43:9–12. [PubMed: 26922774]
37. Hjalgrim H, et al. No evidence of transmission of chronic lymphocytic leukemia through blood
Author Manuscript
Nat Rev Dis Primers. Author manuscript; available in PMC 2017 March 04.
Kipps et al. Page 27
48. Ferreira PG, et al. Transcriptome characterization by RNA sequencing identifies a major molecular
and clinical subdivision in chronic lymphocytic leukemia. Genome Res. 2014; 24:212–226.
Author Manuscript
[PubMed: 24265505]
49. Quesada V, et al. Exome sequencing identifies recurrent mutations of the splicing factor SF3B1
gene in chronic lymphocytic leukemia. Nat. Genet. 2012; 44:47–52.
50. Te Raa GD, et al. The impact of SF3B1 mutations in CLL on the DNA-damage response.
Leukemia. 2015; 29:1133–1142. [PubMed: 25371178]
51. Landau DA, et al. Mutations driving CLL and their evolution in progression and relapse. Nature.
2015; 526:525–530. [PubMed: 26466571]
52. Puente XS, et al. Non-coding recurrent mutations in chronic lymphocytic leukaemia. Nature. 2015;
526:519–524. [PubMed: 26200345] References 51 and 52 describe landmark studies on whole-
exome sequencing of CLL cells obtained from a large cohort of patients, reporting new driver
mutations in CLL.
53. Ljungstrom V, et al. Whole-exome sequencing in relapsing chronic lymphocytic leukemia: clinical
impact of recurrent RPS15 mutations. Blood. 2016; 127:1007–1016. [PubMed: 26675346]
54. O’Brien P, Morin P Jr, Ouellette RJ, Robichaud GA. The Pax-5 gene: a pluripotent regulator of B-
Author Manuscript
cell differentiation and cancer disease. Cancer Res. 2011; 71:7345–7350. [PubMed: 22127921]
55. Villamor N, et al. NOTCH1 mutations identify a genetic subgroup of chronic lymphocytic
leukemia patients with high risk of transformation and poor outcome. Leukemia. 2013; 27:1100–
1106. [PubMed: 23295735]
56. Guieze R, et al. Presence of multiple recurrent mutations confers poor trial outcome of relapsed/
refractory CLL. Blood. 2015; 126:2110–2117. [PubMed: 26316624]
57. Wang J, et al. Tumor evolutionary directed graphs and the history of chronic lymphocytic
leukemia. eLife. 2014; 3:e02869.
58. Burger JA, et al. Clonal evolution in patients with chronic lymphocytic leukaemia developing
resistance to BTK inhibition. Nat. Commun. 2016; 7:11589. [PubMed: 27199251]
59. Calin GA, et al. Frequent deletions and down-regulation of micro-RNA genes miR15 and miR16 at
13q14 in chronic lymphocytic leukemia. Proc. Natl Acad. Sci. USA. 2002; 99:15524–15529.
[PubMed: 12434020] This is the seminal description of the involvement of miRNA in any human
disease. This study describes the most common genetic lesion in CLL, namely, the deletion of two
Author Manuscript
closely linked mi RNAs, mir-15-1 and mir-16a, which are commonly downregulated in CLL.
60. Fabbri M, et al. Association of a microRNA/TP53 feedback circuitry with pathogenesis and
outcome of B-cell chronic lymphocytic leukemia. JAMA. 2011; 305:59–67. [PubMed: 21205967]
61. Mraz M, Kipps TJ. MicroRNAs and B cell receptor signaling in chronic lymphocytic leukemia.
Leuk. Lymphoma. 2013; 54:1836–1839. [PubMed: 23597135]
62. Balatti V, et al. TCL1 targeting miR-3676 is codeleted with tumor protein p53 in chronic
lymphocytic leukemia. Proc. Natl Acad. Sci. USA. 2015; 112:2169–2174. [PubMed: 25646413]
63. Bichi R, et al. Human chronic lymphocytic leukemia modeled in mouse by targeted TCL1
expression. Proc. Natl Acad. Sci. USA. 2002; 99:6955–6960. [PubMed: 12011454]
64. Costinean S, et al. Pre-B cell proliferation and lymphoblastic leukemia/high-grade lymphoma in
Eµ-miR155 transgenic mice. Proc. Natl Acad. Sci. USA. 2006; 103:7024–7029. [PubMed:
16641092]
65. Cui B, et al. MicroRNA-155 influences B-cell receptor signaling and associates with aggressive
disease in chronic lymphocytic leukemia. Blood. 2014; 124:546–554. [PubMed: 24914134]
66. Cahill N, et al. 450K–array analysis of chronic lymphocytic leukemia cells reveals global DNA
Author Manuscript
methylation to be relatively stable over time and similar in resting and proliferative compartments.
Leukemia. 2013; 27:150–158. [PubMed: 22922567]
67. Wahlfors J, et al. Genomic hypomethylation in human chronic lymphocytic leukemia. Blood. 1992;
80:2074–2080. [PubMed: 1382719]
68. Ziller MJ, et al. Charting a dynamic DNA methylation landscape of the human genome. Nature.
2013; 500:477–481. [PubMed: 23925113]
69. Kulis M, et al. Epigenomic analysis detects widespread gene-body DNA hypomethylation in
chronic lymphocytic leukemia. Nat. Genet. 2012; 44:1236–1242. [PubMed: 23064414] This paper
describes an analysis of DNA methylation in CLL cases, showing that the two molecular subtypes
Nat Rev Dis Primers. Author manuscript; available in PMC 2017 March 04.
Kipps et al. Page 28
of CLL have differing DNA methylomes that seem to represent epigenetic imprints from distinct
normal B cell subpopulations.
Author Manuscript
70. Landau DA, et al. Locally disordered methylation forms the basis of intratumor methylome
variation in chronic lymphocytic leukemia. Cancer Cell. 2014; 26:813–825. [PubMed: 25490447]
71. Oakes CC, et al. Evolution of DNA methylation is linked to genetic aberrations in chronic
lymphocytic leukemia. Cancer Discov. 2014; 4:348–361. [PubMed: 24356097]
72. Pei L, et al. Genome-wide DNA methylation analysis reveals novel epigenetic changes in chronic
lymphocytic leukemia. Epigenetics. 2012; 7:567–578. [PubMed: 22534504]
73. Queiros AC, et al. A B-cell epigenetic signature defines three biologic subgroups of chronic
lymphocytic leukemia with clinical impact. Leukemia. 2015; 29:598–605. [PubMed: 25151957]
74. Bhoi S, et al. Prognostic impact of epigenetic classification in chronic lymphocytic leukemia: the
case of subset #2. Epigenetics. 2016; 11:449–455. [PubMed: 27128508]
75. Oakes CC, et al. DNA methylation dynamics during B cell maturation underlie a continuum of
disease phenotypes in chronic lymphocytic leukemia. Nat. Genet. 2016; 48:253–264. [PubMed:
26780610]
76. Lam KP, Kuhn R, Rajewsky K. In vivo ablation of surface immunoglobulin on mature B cells by
Author Manuscript
inducible gene targeting results in rapid cell death. Cell. 1997; 90:1073–1083. [PubMed: 9323135]
77. Herishanu Y, et al. The lymph node microenvironment promotes B-cell receptor signaling, NF-
kappaB activation, and tumor proliferation in chronic lymphocytic leukemia. Blood. 2011;
117:563–574. [PubMed: 20940416] This paper describes a study of gene expression profiles of
CLL cells from the blood and lymphoid tissues (blood, marrow and lymph node) of the same
patients with CLL, showing differences in gene expression for cells in the lymph node versus cells
in the blood of the same patient. The lymph node was identified as the crucial site for BCR and
NF-κB activation, and this was more evident in CLL cells that expressed unmutated IGHV.
78. Mockridge CI, et al. Reversible anergy of sIgM-mediated signaling in the two subsets of CLL
defined by VH-gene mutational status. Blood. 2007; 109:4424–4431. [PubMed: 17255355]
79. Duhren-von Minden M, et al. Chronic lymphocytic leukaemia is driven by antigen-independent
cell-autonomous signalling. Nature. 2012; 489:309–312. [PubMed: 22885698]
80. Awan FT, Byrd JC. New strategies in chronic lymphocytic leukemia: shifting treatment paradigms.
Clin. Cancer Res. 2014; 20:5869–5874. [PubMed: 25294898]
Author Manuscript
81. Yarkoni Y, Getahun A, Cambier JC. Molecular underpinning of B-cell anergy. Immunol. Rev.
2010; 237:249–263. [PubMed: 20727040]
82. Getahun A, Beavers NA, Larson SR, Shlomchik MJ, Cambier JC. Continuous inhibitory signaling
by both SHP-1 and SHIP-1 pathways is required to maintain unresponsiveness of anergic B cells.
J. Exp. Med. 2016; 213:751–769. [PubMed: 27114609]
83. O’Neill SK, et al. Monophosphorylation of CD79a and CD79b ITAM motifs initiates a SHIP-1
phosphatase-mediated inhibitory signaling cascade required for B cell anergy. Immunity. 2011;
35:746–756. [PubMed: 22078222]
84. Packham G, et al. The outcome of B-cell receptor signaling in chronic lymphocytic leukemia:
proliferation or anergy. Haematologica. 2014; 99:1138–1148. [PubMed: 24986876]
85. Cambier JC, Gauld SB, Merrell KT, Vilen BJ. B-cell anergy: from transgenic models to naturally
occurring anergic B cells? Nat. Rev. Immunol. 2007; 7:633–643. [PubMed: 17641666]
86. de Rooij MF, 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;
119:2590–2594. [PubMed: 22279054]
Author Manuscript
87. Vlad A, et al. Down-regulation of CXCR4 and CD62L in chronic lymphocytic leukemia cells is
triggered by B-cell receptor ligation and associated with progressive disease. Cancer Res. 2009;
69:6387–6395. [PubMed: 19654311]
88. Quiroga MP, et al. B-cell antigen receptor signaling enhances chronic lymphocytic leukemia cell
migration and survival: specific targeting with a novel spleen tyrosine kinase inhibitor, R406.
Blood. 2009; 114:1029–1037. [PubMed: 19491390]
89. O’Hayre M, et al. Elucidating the CXCL12/CXCR4 signaling network in chronic lymphocytic
leukemia through phosphoproteomics analysis. PLoS ONE. 2010; 5:e11716. [PubMed: 20661426]
Nat Rev Dis Primers. Author manuscript; available in PMC 2017 March 04.
Kipps et al. Page 29
90. Endo T, et al. BAFF and APRIL support chronic lymphocytic leukemia B-cell survival through
activation of the canonical NF-kappaB pathway. Blood. 2007; 109:703–710. [PubMed: 16973958]
Author Manuscript
91. Aguilar-Hernandez MM, et al. IL-4 enhances expression and function of surface IgM in CLL cells.
Blood. 2016; 127:3015–3025. [PubMed: 27002119]
92. Lu D, et al. Activation of the Wnt signaling pathway in chronic lymphocytic leukemia. Proc. Natl
Acad. Sci. USA. 2004; 101:3118–3123. [PubMed: 14973184]
93. Yu J, et al. Wnt5a induces ROR1/ROR2 heterooligomerization to enhance leukemia chemotaxis
and proliferation. J. Clin. Invest. 2016; 126:585–598. [PubMed: 26690702]
94. Cui B, et al. High-level ROR1 associates with accelerated disease-progression in chronic
lymphocytic leukemia. Blood. 2016; 128:2931–2940. [PubMed: 27815263]
95. Seke Etet PF, Vecchio L, Nwabo Kamdje AH. Interactions between bone marrow stromal
microenvironment and B-chronic lymphocytic leukemia cells: any role for Notch, Wnt and Hh
signaling pathways? Cell Signal. 2012; 24:1433–1443. [PubMed: 22446006]
96. Decker S, et al. Trisomy 12 and elevated GLI1 and PTCH1 transcript levels are biomarkers for
Hedgehog-inhibitor responsiveness in CLL. Blood. 2012; 119:997–1007. [PubMed: 22130798]
97. De Falco F, et al. Notch signaling sustains the expression of Mcl-1 and the activity of eIF4E to
Author Manuscript
21968788]
102. Pepper C, et al. Phenotypic heterogeneity in IGHV-mutated CLL patients has prognostic impact
and identifies a subset with increased sensitivity to BTK and PI3Kdelta inhibition. Leukemia.
2015; 29:744–747. [PubMed: 25349153]
103. Niemann CU, et al. Disruption of in vivo chronic lymphocytic leukemia tumor-microenvironment
interactions by ibrutinib — findings from an investigator-initiated phase II study. Clin. Cancer
Res. 2016; 22:1572–1582. [PubMed: 26660519]
104. Izcue A, Coombes JL, Powrie F. Regulatory T cells suppress systemic and mucosal immune
activation to control intestinal inflammation. Immunol. Rev. 2006; 212:256–271. [PubMed:
16903919]
105. DiLillo DJ, et al. Chronic lymphocytic leukemia and regulatory B cells share IL-10 competence
and immunosuppressive function. Leukemia. 2013; 27:170–182. [PubMed: 22713648]
106. Ramsay AG, Clear AJ, Fatah R, Gribben JG. Multiple inhibitory ligands induce impaired T-cell
immunologic synapse function in chronic lymphocytic leukemia that can be blocked with
lenalidomide: establishing a reversible immune evasion mechanism in human cancer. Blood.
Author Manuscript
Nat Rev Dis Primers. Author manuscript; available in PMC 2017 March 04.
Kipps et al. Page 30
109. Alapat D, et al. Diagnostic usefulness and prognostic impact of CD200 expression in lymphoid
malignancies and plasma cell myeloma. Am. J. Clin. Pathol. 2012; 137:93–100. [PubMed:
Author Manuscript
22180482]
110. Fukuda T, et al. Antisera induced by infusions of autologous Ad-CD154-leukemia B cells identify
ROR1 as an oncofetal antigen and receptor for Wnt5a. Proc. Natl Acad. Sci. USA. 2008;
105:3047–3052. [PubMed: 18287027]
111. Dearden C. B- and T-cell prolymphocytic leukemia: antibody approaches. Hematol. Am. Soc.
Hematol. Educ. Program. 2012; 2012:645–651.
112. Montserrat E, Rozman C. Bone marrow biopsy in chronic lymphocytic leukaemia: a study of 208
cases. Haematologia (Budap.). 1983; 16:73–79. [PubMed: 6679490]
113. Gupta D, Lim MS, Medeiros LJ, Elenitoba-Johnson KS. Small lymphocytic lymphoma with
perifollicular, marginal zone, or interfollicular distribution. Mod. Pathol. 2000; 13:1161–1166.
[PubMed: 11106071]
114. Rai KR, et al. Clinical staging of chronic lymphocytic leukemia. Blood. 1975; 46:219–234.
[PubMed: 1139039]
115. Binet JL, et al. A clinical staging system for chronic lymphocytic leukemia: prognostic
Author Manuscript
127. Byrd JC, 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. [PubMed: 16344317]
128. Eichhorst B, et al. Chronic lymphocytic leukaemia: ESMO clinical practice guidelines for
diagnosis, treatment and follow-up. Ann. Oncol. 2015; 26(Suppl. 5):v78–v84. [PubMed:
26314781]
129. Eichhorst B, et al. Appendix 6: chronic lymphocytic leukaemia: eUpdate published online
September 2016 http://www.esmo.org/Guidelines/Haematological-Malignancies. Ann. Oncol.
2016; 27(Suppl. 5):v143–v144. [PubMed: 27664254]
Nat Rev Dis Primers. Author manuscript; available in PMC 2017 March 04.
Kipps et al. Page 31
130. Robak T. Therapy of chronic lymphocytic leukaemia with purine nucleoside analogues: facts and
controversies. Drugs Aging. 2005; 22:983–1012. [PubMed: 16363884]
Author Manuscript
131. Chang JE, Kahl BS. Bendamustine for treatment of chronic lymphocytic leukemia. Expert Opin.
Pharmacother. 2012; 13:1495–1505. [PubMed: 22663160]
132. Lukenbill J, Kalaycio M. Fludarabine: a review of the clear benefits and potential harms. Leuk.
Res. 2013; 37:986–994. [PubMed: 23787174]
133. Hallek M, et al. Addition of rituximab to fludarabine and cyclophosphamide in patients with
chronic lymphocytic leukaemia: a randomised, open-label, phase 3 trial. Lancet. 2010;
376:1164–1174. [PubMed: 20888994]
134. Goede V, et al. Obinutuzumab plus chlorambucil in patients with CLL and coexisting conditions.
N. Engl. J. Med. 2014; 370:1101–1110. [PubMed: 24401022]
135. Hillmen P, et al. Chlorambucil plus ofatumumab versus chlorambucil alone in previously
untreated patients with chronic lymphocytic leukaemia (COMPLEMENT 1): a randomised,
multicentre, open-label phase 3 trial. Lancet. 2015; 385:1873–1883. [PubMed: 25882396]
136. Wierda WG, et al. Ofatumumab as single-agent CD20 immunotherapy in fludarabine-refractory
chronic lymphocytic leukemia. J. Clin. Oncol. 2010; 28:1749–1755. [PubMed: 20194866]
Author Manuscript
137. Gentile M, et al. Combination of bendamustine and rituximab as front-line therapy for patients
with chronic lymphocytic leukaemia: multicenter, retrospective clinical practice experience with
279 cases outside of controlled clinical trials. Eur. J. Cancer. 2016; 60:154–165. [PubMed:
27127905]
138. Brown JR, et al. Obinutuzumab plus fludarabine/ cyclophosphamide or bendamustine in the
initial therapy of CLL patients: the phase 1b GALTON trial. Blood. 2015; 125:2779–2785.
[PubMed: 25769620]
139. Eichhorst B, et al. First-line chemoimmunotherapy with bendamustine and rituximab versus
fludarabine, cyclophosphamide, and rituximab in patients with advanced chronic lymphocytic
leukaemia (CLL10): an international, open-label, randomised, phase 3, non-inferiority trial.
Lancet Oncol. 2016; 17:928–942. [PubMed: 27216274]
140. Fischer K, et al. Long-term remissions after FCR chemoimmunotherapy in previously untreated
patients with CLL: updated results of the CLL8 trial. Blood. 2016; 127:208–215. [PubMed:
26486789]
Author Manuscript
141. Rossi D, et al. Molecular prediction of durable remission after first-line fludarabine-
cyclophosphamide-rituximab in chronic lymphocytic leukemia. Blood. 2015; 126:1921–1924.
[PubMed: 26276669]
142. Thompson PA, et al. Fludarabine, cyclophosphamide, and rituximab treatment achieves long-term
disease-free survival in IGHV-mutated chronic lymphocytic leukemia. Blood. 2016; 127:303–
309. [PubMed: 26492934] This paper reports an important finding from the seminal clinical trial
of fludarabine, cyclophosphamide and rituximab for the treatment of patients with CLL,
demonstrating long-term disease-free survival following therapy for many patients who had CLL
cells that express mutated IGHV. This study, along with references 139 and 140, shows that a
large proportion of patients with CLL cells that express mutated IGHV potentially may be
‘cured’ by chemoimmunotherapy.
143. Ponader S, 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; 119:1182–1189.
[PubMed: 22180443]
144. Guo A, et al. Heightened BTK-dependent cell proliferation in unmutated chronic lymphocytic
Author Manuscript
Nat Rev Dis Primers. Author manuscript; available in PMC 2017 March 04.
Kipps et al. Page 32
146. Burger JA, et al. Ibrutinib as initial therapy for patients with chronic lymphocytic leukemia. N.
Engl. J. Med. 2015; 373:2425–2437. [PubMed: 26639149] This paper reports on the randomized
Author Manuscript
study of ibrutinib versus chlorambucil in the initial therapy of patients with CLL that showed that
ibrutinib was more effective than chemotherapy in the treatment of patients, resulting in the
regulatory agency approval of ibrutinib for the initial treatment of patients with CLL.
147. Woyach JA, et al. Resistance mechanisms for the Bruton’s tyrosine kinase inhibitor ibrutinib. N.
Engl. J. Med. 2014; 370:2286–2294. [PubMed: 24869598] This paper reports on genetic studies
of CLL cells from patients who developed resistance to ibrutinib, showing that a large subset of
such patients had mutations in BTK, which encodes the enzyme inhibited by ibrutinib. As such,
this study demonstrates that inhibition of BTK is responsible for the clinical activity of ibrutinib.
148. Byrd JC, et al. Acalabrutinib (ACP-196) in relapsed chronic lymphocytic leukemia. N. Engl. J.
Med. 2016; 374:323–332. [PubMed: 26641137]
149. Walter HS, et al. A phase 1 clinical trial of the selective BTK inhibitor ONO/GS-4059 in relapsed
and refractory mature B-cell malignancies. Blood. 2016; 127:411–419. [PubMed: 26542378]
150. Wu J, Liu C, Tsui ST, Liu D. Second-generation inhibitors of Bruton tyrosine kinase. J. Hematol.
Oncol. 2016; 9:80. [PubMed: 27590878]
Author Manuscript
151. Blunt MD, Steele AJ. Pharmacological targeting of PI3K isoforms as a therapeutic strategy in
chronic lymphocytic leukaemia. Leuk. Res. Rep. 2015; 4:60–63. [PubMed: 26500849]
152. Furman RR, et al. Idelalisib and rituximab in relapsed chronic lymphocytic leukemia. N. Engl. J.
Med. 2014; 370:997–1007. [PubMed: 24450857]
153. Lampson BL, et al. Idelalisib given front-line for treatment of chronic lymphocytic leukemia
causes frequent immune-mediated hepatotoxicity. Blood. 2016; 128:195–203. [PubMed:
27247136]
154. Coutre SE, et al. Management of adverse events associated with idelalisib treatment: expert panel
opinion. Leuk. Lymphoma. 2015; 56:2779–2786. [PubMed: 25726955]
155. Buensalido JA, Chandrasekar PH. Prophylaxis against hepatitis B reactivation among patients
with lymphoma receiving rituximab. Expert Rev. Anti Infect. Ther. 2014; 12:151–154. [PubMed:
24341369]
156. Friedberg JW, et al. Inhibition of Syk with fostamatinib disodium has significant clinical activity
in non-Hodgkin lymphoma and chronic lymphocytic leukemia. Blood. 2010; 115:2578–2585.
[PubMed: 19965662]
Author Manuscript
157. Souers AJ, et al. ABT-199, a potent and selective BCL-2 inhibitor, achieves antitumor activity
while sparing platelets. Nat. Med. 2013; 19:202–208. [PubMed: 23291630]
158. Del Gaizo Moore V, et al. Chronic lymphocytic leukemia requires BCL2 to sequester prodeath
BIM, explaining sensitivity to BCL2 antagonist ABT-737. J. Clin. Invest. 2007; 117:112–121.
[PubMed: 17200714]
159. Roberts AW, et al. Targeting BCL2 with venetoclax in relapsed chronic lymphocytic leukemia. N.
Engl. J. Med. 2016; 374:311–322. [PubMed: 26639348] This paper reports on seminal clinical
studies of venetoclax, a BCL-2 inhibitor, showing that this drug can be highly effective in the
treatment of patients with CLL, resulting in the initial regulatory agency approval of venetoclax
for the treatment of patients with relapsed CLL with del(17p).
160. Stilgenbauer S, et al. Venetoclax in relapsed or refractory chronic lymphocytic leukaemia with
17p deletion: a multicentre, open-label, phase 2 study. Lancet Oncol. 2016; 17:768–778.
[PubMed: 27178240]
161. Cervantes-Gomez F, et al. Pharmacological and protein profiling suggests venetoclax (ABT-199)
Author Manuscript
as optimal partner with ibrutinib in chronic lymphocytic leukemia. Clin. Cancer Res. 2015;
21:3705–3715. [PubMed: 25829398]
162. Thijssen R, et al. Resistance to ABT-199 induced by microenvironmental signals in chronic
lymphocytic leukemia can be counteracted by CD20 antibodies or kinase inhibitors.
Haematologica. 2015; 100:e302–e306. [PubMed: 25957396]
163. Thompson PA, Wierda WG. Eliminating minimal residual disease as a therapeutic end point:
working toward cure for patients with CLL. Blood. 2016; 127:279–286. [PubMed: 26576865]
Nat Rev Dis Primers. Author manuscript; available in PMC 2017 March 04.
Kipps et al. Page 33
164. Castro JE, Sandoval-Sus JD, Bole J, Rassenti L, Kipps TJ. Rituximab in combination with high-
dose methylprednisolone for the treatment of fludarabine refractory high-risk chronic
Author Manuscript
169. Shanafelt TD, et al. Quality of life in chronic lymphocytic leukemia: an international survey of
1482 patients. Br. J. Haematol. 2007; 139:255–264. [PubMed: 17897301]
170. Forconi F, Moss P. Perturbation of the normal immune system in patients with CLL. Blood. 2015;
126:573–581. [PubMed: 26084672]
171. No authors listed.] Intravenous immunoglobulin for the prevention of infection in chronic
lymphocytic leukemia. A Randomized, Controlled Clinical Trial Cooperative Group for the study
of immunoglobulin in chronic lymphocytic leukemia. N. Engl. J. Med. 1988; 319:902–907.
[PubMed: 2901668]
172. Spadaro G, Pecoraro A, De Renzo A, Della Pepa R, Genovese A. Intravenous versus
subcutaneous immunoglobulin replacement in secondary hypogammaglobulinemia. Clin.
Immunol. 2016; 166–167:103–104.
173. Dhalla F, et al. Antibody deficiency secondary to chronic lymphocytic leukemia: should patients
be treated with prophylactic replacement immunoglobulin? J. Clin. Immunol. 2014; 34:277–282.
[PubMed: 24557494]
174. Visco C, et al. Autoimmune cytopenias in chronic lymphocytic leukemia. Am. J. Hematol. 2014;
Author Manuscript
180. Maurer C, et al. Effect of first-line treatment on second primary malignancies and Richter’s
transformation in patients with CLL. Leukemia. 2016; 30:2019–2025. [PubMed: 27133817]
181. Bockorny B, Codreanu I, Dasanu CA. Hodgkin lymphoma as Richter transformation in chronic
lymphocytic leukaemia: a retrospective analysis of world literature. Br. J. Haematol. 2012;
156:50–66. [PubMed: 22017478]
182. Rossi D, Gaidano G. Richter syndrome: pathogenesis and management. Semin. Oncol. 2016;
43:311–319. [PubMed: 27040710]
Nat Rev Dis Primers. Author manuscript; available in PMC 2017 March 04.
Kipps et al. Page 34
183. Rossi D, et al. Stereotyped B-cell receptor is an independent risk factor of chronic lymphocytic
leukemia transformation to Richter syndrome. Clin. Cancer Res. 2009; 15:4415–4422. [PubMed:
Author Manuscript
19509140]
184. Mauro FR, et al. Diagnostic and prognostic role of PET/CT in patients with chronic lymphocytic
leukemia and progressive disease. Leukemia. 2015; 29:1360–1365. [PubMed: 25650091]
185. Tambaro FP, et al. Outcomes for patients with chronic lymphocytic leukemia and acute leukemia
or myelodysplastic syndrome. Leukemia. 2016; 30:325–330. [PubMed: 26290497]
186. Chavez JC, et al. Results of a phase II study of lenalidomide and rituximab for refractory/relapsed
chronic lymphocytic leukemia. Leuk. Res. 2016; 47:78–83. [PubMed: 27285853]
187. Fecteau JF, et al. Lenalidomide inhibits the proliferation of CLL cells via a cereblon/p21WAF1/
Cip1-dependent mechanism independent of functional p53. Blood. 2014; 124:1637–1644.
[PubMed: 24990888]
188. Ramsay AG, et al. Chronic lymphocytic leukemia T cells show impaired immunological synapse
formation that can be reversed with an immunomodulating drug. J. Clin. Invest. 2008; 118:2427–
2437. [PubMed: 18551193]
189. Badoux XC, et al. Lenalidomide as initial therapy of elderly patients with chronic lymphocytic
Author Manuscript
195. Khouri IF, et al. Nonmyeloablative allogeneic stem cell transplantation in relapsed/refractory
chronic lymphocytic leukemia: long-term follow-up, prognostic factors, and effect of human
leukocyte histocompatibility antigen subtype on outcome. Cancer. 2011; 117:4679–4688.
[PubMed: 21455998]
196. Dreger P, et al. TP53, SF3B1, and NOTCH1 mutations and outcome of allotransplantation for
chronic lymphocytic leukemia: six-year follow-up of the GCLLSG CLL3X trial. Blood. 2013;
121:3284–3288. [PubMed: 23435461]
197. Porter DL, et al. Chimeric antigen receptor T cells persist and induce sustained remissions in
relapsed refractory chronic lymphocytic leukemia. Sci. Transl Med. 2015; 7:303ra139.
198. Fraietta JA, et al. Ibrutinib enhances chimeric antigen receptor T-cell engraftment and efficacy in
leukemia. Blood. 2016; 127:1117–1127. [PubMed: 26813675]
199. Hudecek M, et al. Receptor affinity and extracellular domain modifications affect tumor
recognition by ROR1-specific chimeric antigen receptor T cells. Clin. Cancer Res. 2013;
19:3153–3164. [PubMed: 23620405]
200. Deniger DC, et al. Sleeping beauty transposition of chimeric antigen receptors targeting receptor
Author Manuscript
tyrosine kinase-like orphan receptor-1 (ROR1) into diverse memory T-cell populations. PLoS
ONE. 2015; 10:e0128151. [PubMed: 26030772]
201. Sharma P, Allison JP. Immune checkpoint targeting in cancer therapy: toward combination
strategies with curative potential. Cell. 2015; 161:205–214. [PubMed: 25860605]
202. McClanahan F, et al. PD-L1 checkpoint blockade prevents immune dysfunction and leukemia
development in a mouse model of chronic lymphocytic leukemia. Blood. 2015; 126:203–211.
[PubMed: 25800048]
203. Yu J, et al. Cirmtuzumab inhibits Wnt5a–induced Rac1-activation in chronic lymphocytic
leukemia treated with ibrutinib. Leukemia. 2016 http://dx.doi.org/10.1038/leu.2016.368.
Nat Rev Dis Primers. Author manuscript; available in PMC 2017 March 04.
Kipps et al. Page 35
204. Strati P, Shanafelt TD. Monoclonal B-cell lymphocytosis and early-stage chronic lymphocytic
leukemia: diagnosis, natural history, and risk stratification. Blood. 2015; 126:454–462. [PubMed:
Author Manuscript
26065657]
205. Shanafelt TD, Ghia P, Lanasa MC, Landgren O, Rawstron AC. Monoclonal B-cell lymphocytosis
(MBL): biology, natural history and clinical management. Leukemia. 2010; 24:512–520.
[PubMed: 20090778]
206. Rawstron AC, et al. Monoclonal B-cell lymphocytosis and chronic lymphocytic leukemia. N.
Engl. J. Med. 2008; 359:575–583. [PubMed: 18687638]
207. Cherepakhin V, Baird SM, Meisenholder GW, Kipps TJ. Common clonal origin of chronic
lymphocytic leukemia and high-grade lymphoma of Richter’s syndrome. Blood. 1993; 82:3141–
3147. [PubMed: 7693038]
208. Chen YA, Wang RC, Yang Y, Chuang SS. Epstein-Barr virus-positive diffuse large B cell
lymphoma arising from a chronic lymphocytic leukemia: overlapping features with classical
Hodgkin lymphoma. Pathol. Int. 2016; 66:393–397. [PubMed: 27170210]
209. Chigrinova E, et al. Tw o main genetic pathways lead to the transformation of chronic
lymphocytic leukemia to Richter syndrome. Blood. 2013; 122:2673–2682. [PubMed: 24004666]
Author Manuscript
210. Rossi D, et al. The genetics of Richter syndrome reveals disease heterogeneity and predicts
survival after transformation. Blood. 2011; 117:3391–3401. [PubMed: 21266718]
211. Fabbri G, et al. Genetic lesions associated with chronic lymphocytic leukemia transformation to
Richter syndrome. J. Exp. Med. 2013; 210:2273–2288. [PubMed: 24127483]
212. Jackson HJ, Rafiq S, Brentjens RJ. Driving CAR T-cells forward. Nat. Rev. Clin. Oncol. 2016;
13:370–383. [PubMed: 27000958]
213. Landau DA, Wu CJ. Chronic lymphocytic leukemia: molecular heterogeneity revealed by high-
throughput genomics. Genome Med. 2013; 5:47. [PubMed: 23731665]
Author Manuscript
Author Manuscript
Nat Rev Dis Primers. Author manuscript; available in PMC 2017 March 04.
Kipps et al. Page 36
Monoclonal B lymphocytosis
Monoclonal B lymphocytosis is defined as <5,000 clonal B cells per µl in the blood
without other signs of lymphoma, such as enlarged lymph nodes (>1.5 cm), which would
Author Manuscript
suggest the diagnosis of small lymphocytic lymphoma204. In most, but not all, cases, the
clonal B cells in monoclonal B lymphocytosis express CD5 and have the same immune
phenotype as CLL205. Although biopsies are not generally performed, the incidental
finding of CLL-like cells in the marrow or in normal-sized lymph nodes does not exclude
the diagnosis of monoclonal B lymphocytosis.
Nat Rev Dis Primers. Author manuscript; available in PMC 2017 March 04.
Kipps et al. Page 37
The lymphoma cells in Richter syndrome are malignant B cells that most often resemble
those of non-germinal centre diffuse large B cell lymphoma (DLBCL), differing
morphologically from the original chronic lymphocytic leukaemia (CLL) population. In
addition, the lymphoma cells of over half the patients with Richter syndrome might not
express CD5 or CD23, which are almost invariably expressed by CLL cells. Nevertheless,
the DLBCL-like lymphoma in Richter syndrome often shares the same IGHV-DJ
rearrangement as the original CLL clone207. As such, the lymphoma cells in Richter
syndrome can express unmutated immunoglobulin heavy-chain variable region gene
(IGHV), unlike de novo DLBCL, which virtually always expresses IGHV with somatic
mutations. However, ~20% of the DLBCL-type Richter syndrome and ~50% of Hodgkin
lymphoma-type Richter syndrome have IGHV-DJ rearrangements that differ from that of
the original CLL clone, suggesting that these lymphomas might represent a de novo
Author Manuscript
negative regulator of cell cycle progression from G1 to S phase, is mutated and/or deleted
in ~30–50% of cases, but rarely so in CLL or de novo DLBCL209,211. Finally, Richter
syndrome lymphomas typically do not have mutations in genes encoding proteins that are
involved in nuclear factor-κB signalling or in the transcriptional repressors PRDM1/
BLIMP1 or BCL6, which are common in de novo DLBCL.
Author Manuscript
Nat Rev Dis Primers. Author manuscript; available in PMC 2017 March 04.
Kipps et al. Page 38
Retrovirus vectors are used to introduce the CAR gene into T cells, which integrates into
the genome of the T cell for stable expression. When the CAR T cells are exposed to the
respective antigen, antigen binding triggers activation and expansion of the CAR T cells
and eliminates the cells with target antigen. The binding domain of the CAR can be
directed against any surface antigen.
Author Manuscript
Author Manuscript
Author Manuscript
Nat Rev Dis Primers. Author manuscript; available in PMC 2017 March 04.
Kipps et al. Page 39
Author Manuscript
Author Manuscript
Author Manuscript
Normal naive B cells that have undergone successful V(D)J recombination and express
functional B cell receptors that are capable of binding to antigen interact with CD4+ T cells
and accessory cells, which aggregate to form follicles that become germinal centres.
Germinal cells each have a dark zone, comprising rapidly dividing B cells, and a light zone,
comprising B cells mixed with follicular dendritic cells (FDCs), macrophages and helper T
cells (TH cells). The B cells enter the dark zone of the germinal centre where they
experience rapid proliferation and somatic hypermutation (SHM) in the genes encoding the
Nat Rev Dis Primers. Author manuscript; available in PMC 2017 March 04.
Kipps et al. Page 40
immunoglobulin variable regions of the heavy chain (IGHV) and the light chain (IGVL). As
Author Manuscript
they pass through to the light zone, the B cells that express the fittest B cell receptors for
binding antigen are selected and may undergo immunoglobulin class-switch recombination.
Chronic lymphocytic leukaemia (CLL) cells that use unmutated IGHV apparently originate
from CD5+ B cells prior to experiencing SHM, whereas CLL cells that use mutated IGHV
most likely originate from CD5+ B cells that have passed through and differentiated in the
germinal centre. Some CLL cells might be derived from B cells that also have undergone
immunoglobulin class-switch recombination and express immunoglobulin isotypes other
than IgM and IgD, for example, IgG or IgA. Another subset is one with CLL cells that
express immunoglobulin with only modest somatic mutations, such as CLL cells that use
IGHV3-21 with ~97% homology to the inherited IGHV3-21 gene and an immunoglobulin
light chain encoded by an unmutated IGLV3-21; these cells might derive from a B cell that
has had constrained SHM, possibly owing to a limited need for immunoglobulin somatic
Author Manuscript
Nat Rev Dis Primers. Author manuscript; available in PMC 2017 March 04.
Kipps et al. Page 41
Author Manuscript
Author Manuscript
Author Manuscript
Nat Rev Dis Primers. Author manuscript; available in PMC 2017 March 04.
Kipps et al. Page 42
growth response 2; ELF4, ETS-related transcription factor Elf-4; ERK, extracellular signal-
Author Manuscript
regulated kinase; EWSR1, Ewing sarcoma breakpoint region 1 protein; FBXW7, F-box/WD
repeat-containing protein 7; FUBP1, far upstream element-binding protein 1; GNB1,
guanine nucleotide-binding protein G(I)/G(S)/G(T) subunit β1; H3K4, histone H3 lysine 4;
IC, intracellular domain; IKZF3, Ikaros family zinc-finger protein 3; IL-1R, IL-1 receptor;
IRF4, interferon regulatory factor 4; ITPKB, inositol-trisphosphate 3-kinase B; LRP, low-
density lipoprotein receptor-related protein; MAP2K1, dual-specificity mitogen-activated
protein kinase kinase 1; MAPK, mitogen-activated protein kinase; MED12, Mediator of
RNA polymerase II transcription subunit 12; MGA, MAX gene-associated protein; MYD88,
myeloid differentiation primary response protein MyD88; NF-κB, nuclear factor-κB; NXF1,
nuclear RNA export factor 1; P, phosphate; POT1, protection of telomeres protein 1;
PTPN11, tyrosine-protein phosphatase non-receptor type 11; RIPK1, receptor-interacting
serine/threonine-protein kinase 1; RPS15, 40S ribosomal protein S15; SAMHD1, SAM
Author Manuscript
Nat Rev Dis Primers. Author manuscript; available in PMC 2017 March 04.
Kipps et al. Page 43
Author Manuscript
Author Manuscript
Author Manuscript
phosphorylation (mainly LYN) of CD79A and CD79B that creates a docking site for the
binding and activation of spleen tyrosine kinase (SYK). SYK then triggers the formation of a
multi-component ‘signalosome’, comprising Bruton tyrosine kinase (BTK), AKT,
phosphoinositide 3-kinase (PI3K), phospholipase Cγ2 (PLCγ2) and B cell-linker protein
(BLNK), among others. CD19 is a co-receptor for BCR and is important for PI3K activation,
which recruits and activates PLCγ2, BTK and AKT. PLCγ2 generates diacylglycerol (DAG)
and inositol-1,4,5-trisphosphate (Ins(1,4,5)P3), which triggers Ca2+ release from the
endoplasmic reticulum, leading to the activation of the MEK–extracellular signal-regulated
Nat Rev Dis Primers. Author manuscript; available in PMC 2017 March 04.
Kipps et al. Page 44
kinase (ERK) and nuclear factor-κB (NF-κB) signalling pathways. Other effects of BCR
Author Manuscript
Nat Rev Dis Primers. Author manuscript; available in PMC 2017 March 04.
Kipps et al. Page 45
Author Manuscript
Author Manuscript
Author Manuscript
ligand (APRIL; also known as TNFSF13). In addition, CLL cell survival can be promoted
through cognate interactions between CD31 and CD38, and the production by stromal cells
of WNT factors, which can interact with ROR1, ROR2 and/or various Frizzled receptors.
CLL cell contact with mesenchymal stromal cells can also be established through vascular
cell adhesion protein 1 (VCAM1)– α4β 1 integrin interactions that contribute to CLL cell
survival. In turn, CLL cells can secrete chemokines, such as CCL3 and CCL4, which can
recruit T cells and NLC-precursor cells (monocytes) to the CLL microenvironment.
Activated T cells can provide CLL cells with proliferative signals through CD40 ligand
Nat Rev Dis Primers. Author manuscript; available in PMC 2017 March 04.
Kipps et al. Page 46
(CD40L)-CD40 interactions and the secretion of several cytokines, such as IL-2, IL-4 and
Author Manuscript
IL-10. In turn, activated CLL cells secrete CCL12 and CCL22, which attract more T cells
into the CLL microenvironment. In tissues, CLL cells can be exposed to environmental
and/or self-antigens that might trigger B cell activation through interactions with the surface
immunoglobulin; this could amplify the responsiveness of CLL cells to the signals and
factors that are provided by the CLL microenvironment. BAFFR, BAFF receptor (also
known as TNFRSF13C); BCMA, B cell maturation protein (also known as TNFRSF17);
BCR, B cell receptor; TACI, transmembrane activator and CAML interactor (also known as
TNFRSF13B).
Author Manuscript
Author Manuscript
Author Manuscript
Nat Rev Dis Primers. Author manuscript; available in PMC 2017 March 04.
Kipps et al. Page 47
Author Manuscript
Nat Rev Dis Primers. Author manuscript; available in PMC 2017 March 04.
Kipps et al. Page 48
Author Manuscript
Author Manuscript
Nat Rev Dis Primers. Author manuscript; available in PMC 2017 March 04.
Kipps et al. Page 49
Author Manuscript
Author Manuscript
Nat Rev Dis Primers. Author manuscript; available in PMC 2017 March 04.
Kipps et al. Page 50
Author Manuscript
Author Manuscript
Author Manuscript
Indications for therapy of patients with chronic lymphocytic leukaemia (CLL) include late-
stage disease, evidence for rapid disease progression or disease-related symptoms. Patients
with del(17p) or mutated TP53 should be treated with therapy that does not require
functional TP53, such as ibrutinib (a Bruton tyrosine kinase (BTK) inhibitor), given the
relatively poor outcome for such patients with chemotherapy. For patients without del(17p)
or known mutations in TP53, immunoglobulin heavy-chain variable region (IGHV)
mutational status can help to define the treatment strategy; patients with unmutated IGHV
Nat Rev Dis Primers. Author manuscript; available in PMC 2017 March 04.
Kipps et al. Page 51
could be considered for therapy with a BTK inhibitor (such as ibrutinib) and patients with
Author Manuscript
de novo del(17p) or TP53 mutations, or have known del(17p) and/or TP53 mutations, or
who develop resistance or intolerance to ibrutinib, could be considered for therapy with
idelalisib and rituximab or the BCL-2 inhibitor venetoclax. Patients treated with CIT who do
not have del(17p) or TP53 mutations could be considered for repeat CIT if their progression-
free survival after CIT is >2 years and the patient has sufficient myeloid reserve. Such
patients also might be treated with a BTK inhibitor or a phosphoinositide 3-kinase (PI3K)
inhibitor, which also could be considered for patients who develop intolerance or resistance
to therapy with ibrutinib. Patients who develop resistance or intolerance to inhibitors of
BTK, PI3K and/or BCL-2 should be considered for clinical trials or alternative agents. LDT,
lymphocyte doubling time.
Author Manuscript
Author Manuscript
Nat Rev Dis Primers. Author manuscript; available in PMC 2017 March 04.
Kipps et al. Page 52
Table 1
*
These life-expectancy estimates are increasing with the advent of newer therapies.
Author Manuscript
Author Manuscript
Author Manuscript
Nat Rev Dis Primers. Author manuscript; available in PMC 2017 March 04.
Kipps et al. Page 53
Table 2
*
These life-expectancy estimates are increasing with the advent of newer therapies.
‡
There are five sites of lymphoid organs: cervical, axillary and inguinal nodes, the spleen and the liver.
Author Manuscript
Author Manuscript
Author Manuscript
Nat Rev Dis Primers. Author manuscript; available in PMC 2017 March 04.