Transcript
  • 7/30/2019 Leucemias Cronicas Llc1 2010

    1/7

    Novel Insights into the Biology of CLL

    Mark C. Lanasa1

    1Division of Medical Oncology, Duke University Medical Center, Durham NC

    Significant advancements in the care of patients with chronic lymphocytic leukemia (CLL) have occurred over the pastdecade. Nonetheless, CLL remains incurable outside of allogeneic transplantation. CLL is the most common leukemia

    in the United States and Europe, and new treatments and therapeutic strategies are clearly needed. To address this

    need, the pathogenesis of CLL has been an area of intense ongoing investigation. These international efforts illuminate

    a complex biology that is reliant on the interplay of inherited, environmental, and host factors. This broad review will

    discuss the recent advances in our understanding of CLL biology including the elucidation of inherited and acquired

    genetic changes; the role of the B-cell receptor and B-cell receptor signaling; CLL cell kinetics; and the interactions in

    the microenvironment between CLL cells, other immune cells, and stromal elements. This improved understanding of

    disease pathogenesis is facilitating the development of novel therapeutic treatment strategies.

    Chronic lymphocytic leukemia (CLL) remains an enigmatic dis-

    ease. Although the first clinical description of chronic lymphoidleukemia was published over 150 years ago, the etiology of CLL is

    unknown, the cell of origin of CLL is unknown, and CLL remains

    incurable outside of allogeneic transplantation. CLL is the most

    common leukemia in the United States, with approximately 15,500

    new diagnoses per year. Because of the relatively long survival of

    patients with CLL, it is by far the most prevalent leukemia, with an

    estimated 95,000 Americans living with CLL.1 CLL lymphocytes

    have a characteristic immunophenotype: CD5, CD19, CD20dim,

    CD23, and surface immunoglobulindim. Although the majority of

    patients are asymptomatic at diagnosis, the relentless accumulation

    of CLL lymphocytes leads to symptomatic disease, need for

    CLL-directed therapy, disease-related complications, and approxi-

    mately 4500 CLL attributable deaths per year in the United States.

    Several characteristics of CLL facilitate basic and translational

    research: (i) the high population prevalence; (ii) the malignant cells

    are easily obtained through venous phlebotomy; (iii) most patients

    have an asymptomatic phase that allows for longitudinal evaluation;

    and (iv) CLL is has a relatively long disease-specific survival.

    Therefore, CLL has become a model system for the investigation of

    B-cell lymphoproliferative disorders. In the 5 years since the

    biology of CLL was last broadly reviewed in the American Society

    of Hematology education session, tremendous progress has been

    made in the understanding of CLL disease biology, and this review

    will focus on these discoveries. Specifically, important advances

    have been made in identifying inherited and acquired genetic

    mutations, the role of B-cell receptor (BCR) signaling, and the

    interplay between the malignant B cells and the tumor microenviron-ment. These advances reveal CLL to be a disease that is dependent

    on on the interplay of inherited, environmental, and host factors.

    Inherited Genetic FactorsThe most important risk factor for the development of CLL is a

    family history of CLL. Among patients with newly diagnosed CLL,

    8% to 10% have a family history of CLL. CLL has a heritability that

    is twice that of common solid tumors with known low-prevalence,

    high penetrance causative genes, such as breast and colon cancer. 2,3

    Pedigree analysis using data from the Swedish Family Cancer

    Database showed the relative risk of CLL among first-degree

    relatives of persons with CLL to range between 7.0 and 8.5. This

    equivalent risk across all first-degree relatives suggests an inherited,rather than environmental, basis for familial CLL. 4 Though families

    with four or more cases of CLL have been reported, they are

    extremely rare. Some CLL kindreds suggest a dominant inheritance,

    therefore, genome-wide linkage studies in familial CLL have been

    performed. The largest and most recent reported study added 101

    new cases of familial CLL to a previously reported cohort of 105

    families.5 This study was the first statistically significant genome-

    wide linkage scan and identified chromosome 2q21.2 as associated

    with inheritance of CLL. However, no causative genes have been

    identified at this locus. Overall, linkage studies in CLL have been

    limited by the low number of affected individuals per family, late

    age of onset, and presumed genetic heterogeneity.

    Complete sequencing of the human genome revealed large numbers

    of common single nucleotide polymorphisms (SNPs). This discov-

    ery led to the hypothesis that the inheritance of complex diseases

    may be due to the coinheritance of these common variants. Di

    Bernardo et al6 performed a multistage genome-wide SNP associa-

    tion (GWA) study to evaluate the contribution of common variants

    to the inheritance of CLL. This study identified six novel loci

    associated with the development of CLL, thus providing the first

    evidence for the contribution of common (population prevalence

    5%) genetic variants to the development of CLL. Five of these SNPs

    have been validated in an independent GWA.7 Subsequently, four

    additional susceptibility loci have been identified through follow-up

    analyses from the initial Di Bernardo study.8 Identification of these

    genes informs our understanding of CLL biology. For example, theGWA linked interferon regulatory factor 4 (IRF4), a key regulator

    of lymphocyte maturation and proliferation, with risk of developing

    CLL. The 10 loci identified to date individually confer a small risk

    of disease, with relative risks ranging from 1.2 to 1.6. As a group,

    these risk alleles account for 10% of the total heritability of CLL.

    Because GWA can detect only those alleles with a population

    prevalence of 5%, it is possible that much of the inherited risk of

    CLL is due to uncommon inherited variants (prevalence 5%).

    Massively parallel sequencing technologies may enable the discov-

    ery of these rare variants and illuminate the hidden heritability

    in CLL.

    CHRONIC LYMPHOCYTIC LEUKEMIA

    70 American Society of Hematology

  • 7/30/2019 Leucemias Cronicas Llc1 2010

    2/7

    Acquired Genetic FactorsIn a landmark report by the German CLL Study Group, Dohner et

    al9 showed that acquired chromosomal abnormalities involving

    chromosomes 11, 12, 13, and 17 are common in CLL, and that these

    abnormalities predict both time to first treatment and CLL-specific

    survival. The genes principally responsible for the adverse progno-

    sis associated with del 17p13 and del 11q22 were recognized to be

    TP53 and ATM, respectively. The genes that contribute to CLL

    pathogenesis in trisomy 12 remain unknown. There is ongoing

    debate regarding whether trisomy 12 confers an increased risk of

    disease progression; unlike the Dohner study, our institutional

    experience is that patients with trisomy 12 do follow a somewhat

    more aggressive disease course than those patients with normal

    FISH (fluorescence in situ hybridization).10 Deletion of 13q14 is the

    most common and most favorable cytogenetic abnormality in CLL.

    The responsible genes at this locus were initially unclear. In 2002,

    Calin et al11 showed that the microRNA 15/16 (miR 15/16) cluster

    were the critical deleted genes in this region. MicroRNAs are small

    (approximately 20 nucleotides) nonprotein coding RNAs that

    modulate the level of specific proteins by binding sequence-

    complimentary mRNAs. miR-15 and miR-16 were subsequently

    shown to be negative regulators of BCL2.12 This discovery is

    notable in that it was the first association of clinical disease with

    microRNAs.

    The Dohner study9 established interphase cytogenetics (FISH) as

    the clinical standard of care for evaluation of chromosomal aberra-

    tions in CLL because the low proliferative capacity of most CLL

    limits the clinical utility of metaphase cytogenetics. However,

    emerging data shows that the evaluation of four (or five) chromo-

    somal loci by FISH obscures the significant heterogeneity and

    complexity of acquired chromosomal defects in CLL. The applica-

    tion of high-density SNP arrays illuminates some of this complex-

    ity. For example, copy number neutral loss of heterozygosity is

    observed in a significant number of CLL cases, and this finding

    cannot be detected with either FISH or standard karyotyping. Using

    SNP arrays, Ouilette et al13 showed significant heterogeneity among

    13q14 deletions, with some deletions extending centromeric to

    include the retinoblastoma gene. A follow-up study showed that

    13q14 deletions that include retinoblastoma are associated with

    genomic complexity,14 a finding associated with aggressive disease

    in prior clinical reports. Although data currently available are

    inadequate to support the widespread application of SNP arrays to

    routine clinical care, this technology may ultimately be a useful

    adjunct to routine clinical FISH.

    At the chromosomal level, del 17p13 is more homogenous than

    other acquired genomic defects. High-density SNP arrays showed

    that the deletion breakpoint typically falls within chromosomal band

    17p11.2 and extends telomeric to include most of the p arm.15

    Deletions of 17p13 are almost always monoallelic, and it was

    initially unclear why monoallelic loss of TP53 would cause such a

    dramatic change in CLL proliferative capacity, chemosensitivity,

    and prognosis. It is now understood that 80% of cases with del

    17p13 have single nucleotide somatic mutations of the TP53 allele

    on the other chromosome 17 (the allele in trans), thereby causing

    near complete loss of TP53 function. Although uncommon, TP53

    inactivating mutations can occur in the absence of del 17p13, and

    these mutations apparently confer an equivalent adverse risk as del

    17p13.16 TP53 is a multimeric protein with four identical subunits,

    and these inactivating mutations may act in a dominant negative

    manner, thus abrogating TP53 function even in the presence of a

    wild-type allele in trans. Although only 5% of CLL cases show

    deletion or mutation of 17p13 at diagnosis, somatic mutations occur

    in approximately one-third of relapsed or chemotherapy refractory

    patients. Whether this represents clonal selection or clonal evolution

    or both is an area of active investigation.

    Epigenetic changes are also relevant to the pathogenesis of CLL.

    Epigenetic changes are noninherited chromosomal modifications

    that affect gene transcription, such as methylation of gene promoters

    and acetylation of histone-bound DNA. An example of epigenetic

    change, global hypomethylation of DNA, has been described in

    CLL. Between 2% and 8% of CpG islands (gene promoter

    elements) are aberrantly methylated in CLL when compared with

    normal B cells, a finding that suggests DNA methylation broadly

    affects the transcriptional profile of CLL. A germline mutation in

    death-associated protein kinase 1 (DAPK1) that segregated with

    risk of CLL was identified by Raval et al18 in a family with multiple

    CLL cases. The authors showed that DAPK1 expression is silenced

    through promoter methylation in the majority of CLL cases,

    suggesting a central role for both epigenetic modification and

    DAPK1 in CLL leukemogenesis.18 Using high-density methylation

    microarrays evaluating over 27,000 CpG sites, Kanduri et al19

    identified differential patterns of methylation that were dependent

    on the mutation status of the BCR. Poor-risk CLL showed

    methylation profiles that facilitated signaling through proliferative

    cellular pathways, including MAPK (mitogen-activated protein

    kinase) and NF-B (nuclear factor- light-chain enhancer of

    activated B cells), a finding that directly relates the clinical

    phenotype of poor-risk CLL to methylation of BCR-mediated

    signaling pathways.

    The Role of BCR in CLLInvestigation of the immunoglobulin gene has led to findings that

    are central to the current understanding of CLL (Table 1). During

    B-cell development, variable-diversity-joining (VDJ) recombina-

    tion generates an immunoglobulin heavy chain (IGH), and an

    immunoglobulin light chain (IGL) is created through isotype-

    specific VJ recombination. There is tremendous combinatorial

    potential in this process, with perhaps 1010 potential IGH-IGL

    sequence combinations. However, patients with CLL use a highly

    restricted set of BCRs.20 Approximately 14% of all CLL cases

    express the heavy chain VH169, and an additional 18% express

    VH434. This finding of marked restriction of the IGH repertoire

    led to the hypothesis that tonic stimulation by specific auto or

    alloantigens drives expansion of the malignant clone.21,22 Intra-

    clonal diversification of some CLL further supports the antigen-

    drive hypothesis.23

    During the germinal center reaction, somatic mutations are gener-

    ated in the BCR to increase affinity for target antigen. Approxi-

    mately half of CLL cases show IGH mutations, and the other half

    Table 1. The mutation status of the immunoglobulin heavy chain (IGVH)

    as a central determinant of CLL biology

    IGVH Unmutated IGVH Mutated

    Cell of origin Mature (antigen experienced) B

    cell

    Mature (antigen experienced) B

    cell

    Postulated mechanism of

    maturation

    Germinal center, T-help

    dependent

    T-help independent, germinal

    center independent (?)

    Chromosomal abnormalities del 11q22, del 17p13 del 13q14 (sole)

    Frequency of BCR stereotypy 40% 10%

    Antigen recognition Polyreactive oligoreactive

    Response to BCR cross-linking Activation, proliferation Anergy or no response

    Hematology 2010 71

  • 7/30/2019 Leucemias Cronicas Llc1 2010

    3/7

    show an unmutated IGH, typically defined as 2% deviation

    from germline sequence. Patients with mutated IGH typically

    follow a more indolent disease course than those with unmutated

    IGH24. This initially led to the hypothesis that CLL may be two

    distinct entities, one deriving from naive (IGH unmutated) B cells

    and the other from postgerminal center memory (IGH mutated) B

    cells. However, gene expression profiling studies in CLL show both

    IGH mutated and unmutated CLL to be derived from memory B

    cells.25,26 The mechanism by which some CLL escape IGH mutation

    is unclear. One hypothesis proposes that certain antigens favor ahelper T-cell independent or germinal center independent matura-

    tion that does not involve IGH mutation. Alternatively, the IGH

    unmutated CLL cells may be autoreactive and targeted for apopto-

    sis, but avoid cell death through either tonic BCR stimulation or

    transforming genetic events.

    Recent experiments using phage display libraries have attempted to

    better define the types of antigens and epitopes recognized by IGH

    mutated and unmutated CLL. Monoclonal antibodies (MAbs)

    generated from the BCR of IGH-mutated CLL bound epitopes with

    structurally related amino acid motifs, whereas unmutated CLL

    recognized multiple different epitopes.27 This suggests that in vivo

    multiple, potentially structurally divergent antigens can bind and

    stimulate CLL cells through the BCR. This conclusion is provoca-

    tive because six of eight of the CLL MAbs investigated had

    stereotyped BCRs. Stereotypy is the observation of near complete

    sequence homology of the complimentarity-determining region 3

    (CDR3) of the IGH28; the CDR3 largely defines the antigen

    specificity of the BCR. For example, approximately 1% of all CLL

    express VH1 69 that are virtually identical within the CDR3.29 The

    presence of a stereotyped BCR is associated with adverse risk,

    independent of IGH mutation status, although stereotypy is more

    commonly observed among unmutated (40%) than mutated CLL

    (10%). The observation of stereotypy has been forwarded as

    evidence of selective antigenic pressure in CLL; however, MAbs

    derived from these BCRs may be oligo- or polyreactive in vivo.

    MAbs derived from stereotyped BCRs have also been shown to

    react with conserved epitopes exposed on apoptotic cells.30 The

    selection of stereotyped BCRs may be related to a combination of

    antigenic pressure and CDR3 sequence facilitated BCR signaling.

    Given the clear importance of the BCR in the clinical behavior of

    CLL, the role of BCR-mediated signaling is another area of

    significant interest. The surface density of the BCR is lower in CLL

    than in most B-cell subsets, suggesting CLL may have attenuated

    signaling responses to antigen binding. Experimental evidence

    shows that, in vitro, approximately half of all CLL retain BCR-

    mediated signaling, and that the IGH mutation status and other

    molecular characteristics used for clinical prognostication (eg,

    CD38 and ZAP70 [zeta-associated protein 70]) are important

    determinants of BCR responsiveness. After cross-linking the BCRs

    with isotype-specific antibodies, Lanham et al31 showed global

    increases in tyrosine phosphorylation, a marker of induction of

    intracellular signaling. The responsiveness to cross-linking was

    strongly associated with IGH mutation status (P .0005), as well as

    expression of CD38 (P .05).31 Using gene expression profiling,

    Guarini et al32 showed that BCR cross-linking with anti-IgM led to

    the transcription of gene pathways regulating cell-cycle regulation,

    cytoskeletal organization, and proliferation; but, these changes were

    only observed among IGH unmutated cases. BCR ligation leads to

    enhanced intracellular signaling among ZAP70 expressing CLL,

    and the introduction of ZAP70 into CLL not otherwise expressing

    ZAP70 augments BCR-mediated signaling, suggesting a direct role

    for ZAP70 in BCR-mediated signaling.33 Muzio et al34 showed that

    CLL B cells that do not respond to BCR ligation (typically

    IGH-mutated cases) show activation of cellular pathways that

    suggest anergy. BCR signaling in vivo is likely even more

    complicated: assuming that there are multiple antigens capable of

    binding the BCR, the binding affinity for a specific antigen likely

    determines whether the response is activating or anergic. Although

    this complexity in vivo remains incompletely understood, in

    general, BCR ligation in IGH unmutated CLL leads to predomi-

    nantly activating and proliferative responses, whereas BCR signal-ing in IGH-mutated CLL favors anergic and antiapoptotic re-

    sponses. Additionally, the adverse risk associated with ZAP70 and

    CD38 expression may be directly related to their effects on cell

    signaling and activation.

    CLL Cell KineticsThe conventional view of CLL had been that it is primarily a disease

    of failed apoptosis and passive accumulation. This view is sup-

    ported by the observation that the great majority ( 98%) of

    peripherally circulating CLL cells are arrested in G0 or the early G1

    phase and have overexpression of antiapoptotic proteins, such as

    BCL2. Other observations suggest that CLL may have significant

    proliferative capacity. As discussed in the prior section, IGH

    unmutated CLL has the capacity for proliferative responses to BCR

    ligation. Many CLL express markers of cellular activation (eg,

    CD38, CD49d, and CD69), providing a link between CLL cell

    biology and prognostics. Clonal evolution and Richters transforma-

    tion are observed in some cases. In a series of elegant experiments,

    Messmer et al 35 evaluated the in vivo kinetics of CLL by having

    patients consume fixed doses of deuterated heavy water (2H2O)

    for 84 days. The deuterium was incorporated into newly synthesized

    DNA during the S phase. Using mass spectrometry, the birth and

    death rates of CLL cells were then determined. Although there

    was significant variability in birth rates (0.11%1.76%/day), all

    patients had a rate of new cell formation of at least 10 9 new CLL

    cells per day. Patients with higher birth rates (exceeding 0.35%/day)

    were more likely to have symptomatic disease. Significant variabil-

    ity was also observed in the cellular death rate, and the balance

    between birth and death likely determines both white blood cell

    trend and risk of progression.35

    The deuterated water experiments also provided novel insights into

    CLL intraclonal heterogeneity and CLL cell trafficking. By flow

    sorting the CD38 and CD38 fractions from CLL patients receiv-

    ing deuterated water, Calissano et al36 showed that the CD38

    fraction proliferated at a greater rate than the CD38 fraction. It has

    previously been shown that, regardless of the proportion of CD38

    cells, these cells show significantly increased expression of prolifera-

    tion factors ZAP70, Ki67, and telomerase when compared with the

    CD38 fraction.37 In a more recent report, Calissano et al provided

    preliminary evidence that new CLL cells have a distinct immuno-

    phenotype: CD5hiCXCR4low, whereas resting CLL cells are

    CD5lowCXCR4hi. Gene expression array analysis of flow sorted

    new and resting cell fractions that showed that new cells

    differentially express genes related to proliferation, cellular activa-

    tion, and cell signaling, whereas resting cells predominantly

    express genes involved in apoptosis, cell death, and migration. 38

    These studies have led to the hypothesis that CLL cells in the blood

    compartment eventually enter a pathway of cellular senescence.

    Upregulation of CXCR4 (a CXC chemokine receptor) promotes

    reentry into the tumor microenvironment, where the cells receive

    prosurvival signals. Rather than undergoing apoptosis, a subset of

    resting CLL cells are activated, proliferate, downregulate CXCR4,

    72 American Society of Hematology

  • 7/30/2019 Leucemias Cronicas Llc1 2010

    4/7

    and are then released back into the peripheral blood. These studies

    shed light on the phenotypic continuum of CLL cells and better

    define the life cycle of a CLL B cell, as well as the central role of the

    tumor microenvironment in maintaining the malignant clone.

    The Tumor Microenvironment in CLLOver the past decade, the essential role of the tumor microenviron-

    ment in the survival and progression of CLL has become increas-

    ingly clear. The tumor microenvironment describes an admixture of

    malignant cells with host immune cells, stromal elements, and

    vascular cells that create a niche wherein signals can be transmitted

    through antigen presentation, cellcell interactions, and paracrine

    signaling. The microenvironments differ in the bone marrow and

    secondary lymphoid organs, where the former contains mesenchy-

    mal stromal cells (MSC) within vascular niches and in the latter

    nurse-like cells (NLCs), T cells and follicular dendritic cells are

    present.

    NLCs are large CD14 mononuclear cells that are abundant in the

    secondary lymphoid organs of patients with CLL.39 NLCs and

    MSCs constitutively express the chemokines CXCL12 (stromal-

    derived factor-1, Sdf-1) and CXCL13. CLL cells express CXCR4

    (CD184), the receptor for Sdf-1, and in vitro studies have shown

    that engagement of Sdf-1 with CXCR4 promotes cell survival

    through activation of the signal transducer and activator of transcrip-

    tion 3 (STAT3) and MAPK pathways. Blockade of this interaction

    with a small molecule inhibitor leads to decreased CLL viability,

    suggesting this interaction is a potential therapeutic target.40 Addi-

    tional studies have shown that CLL cell viability can be significantly

    enhanced in vitro with Sdf-1, although viability is further enhanced

    with NLCs over Sdf-1, suggesting that NLCs also deliver additional

    prosurvival signals. It is now understood that, in addition to Sdf-1,

    NLCs also secrete B-cell activating factor (BAFF) and a proliferation-

    inducing ligand (APRIL). Engagement of these ligands with CLL

    cells induces intracellular NF-B1 and MCL-1, whereas sdf-1

    induces ERK1/2 and AKT. As such, NLCs can deliver multiple

    prosurvival signals that utilize distinct signaling pathways.41 Engage-

    ment of CD40, expressed on the surface of CLL cells, with CD154

    (CD40L), expressed on CD4 T cells found in CLL pseudofollicles,

    also induces the NF-B pathway. These diverse interactions have

    important implications for CLL therapy, particularly as mechanisms

    of chemotherapy resistance. A recent report showed that in vitro

    coculture of CLL cells with CD154 expressing fibroblasts induced

    1000-fold resistance to ABT-737, a small molecular inhibitor of

    BCL2 and BCL-XL.42 Similarly, coculture of CLL cells with MSCs

    induces resistance to fludarabine, cyclophosphamide, and dexameth-

    asone through maintenance of MCL-1.43 Taken together, these in

    vitro data strongly suggest that the tumor microenvironment en-

    hances CLL cell resistance to both apoptosis and chemotherapy.

    Investigation of novel mechanisms of interaction between the tumor

    microenvironment and CLL cells is an active area of research. CLL

    cells have the capacity to affect local angiogenesis within the

    vascular niche. Microvessel density is increased in bone marrow of

    CLL patients, with highest densities found at the periphery of

    lymphoid aggregates. CLL cells have the capacity to secrete a

    variety of angiogenic cytokines, including vascular endothelial

    growth factor (VEGF), basic fibroblast growth factor (bFGF), and

    thrombospondin-1 (TSP-1).44 More recently, Ghosh et al45 de-

    scribed the contribution of CLL cell-derived microvesicles as a

    mechanism by which CLL cells can module the local microenviron-

    ment. Microvesicles are cell membrane-derived particles that can

    deliver cell surface receptors, activated signaling proteins, or nucleic

    acids to target cells. The authors found that CLL cell-derived mi-

    crovesicles were increased in patients with advanced-stage CLL, and

    that microvesicles could activate AKT/mTOR signaling in MSCs.45

    Thus, microvesicles represent a novel mechanism of CLL signaling

    within the microenvironment. Finally, toll-like receptors (TLRs) are

    cell surface receptors that are part of the innate immune system that

    bind structurally conserved microbial antigens and activate immune

    responses. Recent data shows that TLRs are expressed and func-

    tional in CLL; binding of TLRs with cognate antigen-activated

    NF-B expression and induced surface expression of activationmarkers CD25 and CD80.46 Given the architectural complexity,

    number of different cell types present, and clinical importance of

    these interactions, investigation of the tumor microenvironment will

    continue to be an active and fruitful area of research in CLL.

    T-Cell Abnormalities in CLLMost malignancies are associated with decreased numbers of

    circulating T cells, but in CLL they are elevated 2.5 to 4 times

    normal, at least pretherapy.47 Although T cells are increased in

    number, the T-cell repertoire is significantly contracted in CLL,

    with oligoclonal and monoclonal subsets.48,49 CLL cells secrete

    immunomodulatory cytokines, such as interleukin-6, interleukin-

    10, and tumor growth factor- (TGF-), which shift the helper

    T-cell response from a Th1 response to an anergic Th2 response. 50,51

    Suppressive regulatory T cells (Treg) are also increased in patients

    with CLL.52 A mechanism for this increase may be direct CLLT-

    cell contact through CD27 and CD70; this interaction also induces

    BCL2 within the Treg, making the T reg cell population in CLL

    patients more resistant to apoptosis than in normal individuals.53

    These qualitative and quantitative T-cell abnormalities likely allow

    the CLL lymphocytes to avoid cell-mediated immune responses,

    despite the fact that CLL cells express tumor-specific antigens that

    can be presented by major histocompatibility complex molecules.

    To better understand the mechanism underlying impaired T-cell

    responses in CLL, Gorgun et al54 performed gene expression array

    analyses of purified CD4 and CD8 T cells. Among CD4 T cells,

    genes related to cell differentiation were differentially expressed

    between T cells derived from previously untreated CLL patients and

    healthy controls. Gene pathways controlling cytoskeleton forma-

    tion, vesicle trafficking, and cytotoxicity were differentially ex-

    pressed in CD8 T cells. When CLL cells were cocultured with

    CD4 or CD8 T cells obtained from healthy controls, the abnormal

    patterns of gene expression were induced, suggesting that these

    T-cell defects are due to direct cellcell contact. 54 In light of the

    observed dysregulation of cytoskeleton assembly genes, a subse-

    quent study by Ramsay et al55 evaluated the capacity of T cells to

    form a normal immunological synapse. This study showed that T

    cells derived from CLL patients could not form normal cellcell

    contact and that recruitment of T-cell signaling proteins to the

    synapse was blocked. Similarly, coculture of CLL cells with donor

    T cells blocked the formation of an immunologic synapse.55 The

    inability of T cells derived from CLL patients to form a normal

    immunologic synapse likely impairs both antigen recognition and

    cellular cytotoxicity. Taken together, this global impairment in

    T-cell function may be an important cause of tumor progression,

    increased susceptibility to infection, and secondary malignancies in

    patients with CLL.

    Using Novel Biologic Insights to Develop an

    Integrated Model of Disease BiologyDetailed laboratory-based and translational investigation has yielded

    tremendous progress in our understanding of CLL. One of the

    Hematology 2010 73

  • 7/30/2019 Leucemias Cronicas Llc1 2010

    5/7

    significant challenges at this time is to assemble this body of

    knowledge into a unified model of disease pathogenesis. Epidemio-

    logic data implies the importance of ethnicity (likely a marker of

    genetic risk) and age (a marker of immune senescence). A proposed

    model follows: in a genetically susceptible host, tonic stimulation

    by a stereotyped antigenlikely a low-affinity autoantigen

    initiates expansion of a premalignant clone. Through some combina-

    tion of tonic antigenic stimulation, acquisition of somatic genetic

    mutations, and timely support from the tumor microenvironment,

    the potentially autoreactive clone escapes immune surveillance andbegins to expand. In the tumor microenvironment, cells receive

    proliferative signals from CD4 T cells and antiapoptotic signals

    from NLCs and MSCs. Initially supported by the tumor microenvi-

    ronment, the increasing clone may begin to further modulate the

    microenvironment and immune response to favor survival and

    progression of the clone. Among IGH unmutated clones, BCR-

    transmitted signals provide predominantly proliferative signals,

    whereas IGH-mutated clones express gene pathways related to

    cellular anergy. The CLL cells continue to receive antigenic

    stimulation, and as the clone continues to proliferate, additional

    somatic mutations are acquired, and this may diminish the reliance

    of the clone on both antigenic stimulation and microenvironment

    support.

    Some central questions to be investigated include the molecular

    basis of the 2:1 male:female gender bias in CLL, the mechanism by

    which some CLL acquire somatic mutations and others do not (ie,

    the molecular determinants of IGH mutation status), the mechanism

    by which preemergent CLL clones escape immune surveillance and

    deletion, and whether clonal evolution can be avoided through

    suppression of the proliferative compartment of CLL.

    Finally, this scientific progress has created numerous targets of

    novel therapeutic interventions. Lineage-restricted cell surface

    molecules, such as ROR1 and CD37, are targets of novel MAbs.

    Lenalidomide has been shown to reverse the abnormal immunologic

    synapse formation in CLL, and also modulates the microenviron-

    ment through monocyte and NK cell activation. Novel small

    molecule inhibitors of BCR-mediated signaling are currently being

    tested in clinical trials. CAL101 selectively blocks a phosphatidyl-

    inositol 3-kinase isoform related to downstream signaling from

    CXCR4. Additional trials studying small molecular inhibitors of

    antiapoptotic proteins, such as BCL2, are underway. Elucidation of

    the mechanisms of chemotherapy resistance related to loss of p53

    function has led to strategies to circumvent p53-dependent path-

    ways. The advances in our fundamental understanding of the

    mechanisms of disease in CLL will undoubtedly lead to improved

    therapies for our patients.

    DisclosuresConflict-of-interest disclosure: The author has consulted and been

    affiliated with the Speakers Bureau for GlaxoSmithKline and

    Genentech, and has received research funding from Celgene and

    Eleos.

    Off-label drug use: None disclosed.

    CorrespondenceMark C. Lanasa, MD, PhD, Assistant Professor of Medicine,

    Division of Medical Oncology, Duke University Medical Center,

    DUMC Box 3872, 1 Trent Dr., Morris Building Room 25153,

    Durham, NC 27710; Phone: (919) 684-8964; Fax: (919) 684-5325;

    e-mail: [email protected]

    References1. Altekruse SF, Krapcho M, Neyman N, et al. SEER cancer

    statistics review, 19752007. SEER Web site. http://seer.cancer.

    gov/csr/1975_2007/ (based on November 2009 SEER data

    submission, posted to the SEER web site, 2010). Accessed May

    1, 2010.

    2. Amundadottir LT, Thorvaldsson S, Gudbjartsson DF, et al.

    Cancer as a complex phenotype: pattern of cancer distribution

    within and beyond the nuclear family. PLoS Med. 2004;1:e65.

    3. Kerber RA, OBrien E. A cohort study of cancer risk in relationto family histories of cancer in the Utah population database.

    Cancer. 2005;103:1906 1915.

    4. Goldin LR, Pfeiffer RM, Li X, Hemminki K. Familial risk of

    lymphoproliferative tumors in families of patients with chronic

    lymphocytic leukemia: results from the Swedish Family-

    Cancer Database. Blood. 2004;104:1850 1854.

    5. Sellick GS, Wild RW, Slager SL, et al. A high-density SNP

    genome-wide linkage search of 206 families identifies suscepti-

    bility loci for chronic lymphocytic leukemia. Blood. 2007;110:

    33263333.

    6. Di Bernardo MC, Crowther-Swanepoel D, Broderick P, et al. A

    genome-wide association study identifies six susceptibility loci

    for chronic lymphocytic leukemia. Nat Genet. 2008;40(10):

    12041210.

    7. Slager SL, Goldin LR, Strom SS, et al. Genetic susceptibility

    variants for chronic lymphocytic leukemia. Cancer Epidemiol

    Biomarkers Prev. 2010;19:10981102.

    8. Crowther-Swanepoel D, Broderick P, Di Bernardo MC, et al.

    Common variants at 2q37.3, 8q24.21, 15q21.3 and 16q24.1

    influence chronic lymphocytic leukemia risk. Nat Genet. 2010;

    42:132136.

    9. Dohner H, Stilgenbauer S, Benner A, et al. Genomic aberra-

    tions and survival in chronic lymphocytic leukemia. N Engl

    J Med. 2000;343:1910 1916.

    10. Weinberg JB, Volkheimer AD, Chen Y, et al. Clinical and

    molecular predictors of disease severity and survival in chronic

    lymphocytic leukemia. Am J Hematol. 2007;82:10631070.

    11. Calin GA, Dumitru CD, Shimizu M, et al. Frequent deletions

    and down-regulation of micro-RNA genes miR15 and miR16 at

    13q14 in chronic lymphocytic leukemia. Proc Natl Acad Sci

    U S A. 2002;99:15524 15529.

    12. Cimmino A, Calin GA, Fabbri M, et al. miR-15 and miR-16

    induce apoptosis by targeting BCL2. Proc Natl Acad Sci U S A.

    2005;102:1394413949.

    13. Ouillette P, Erba H, Kujawski L, Kaminski M, Shedden K,

    Malek SN. Integrated genomic profiling of chronic lymphocytic

    leukemia identifies subtypes of deletion 13q14. Cancer Res.

    2008;68:10121021.

    14. Ouillette P, Fossum S, Parkin B, et al. Aggressive chronic

    lymphocytic leukemia with elevated genomic complexity is

    associated with multiple gene defects in the response to DNA

    double-strand breaks. Clin Cancer Res. 2010;16:835847.

    15. Fabris S, Mosca L, Todoerti K, et al. Molecular and transcrip-

    tional characterization of 17p loss in B-cell chronic lympho-

    cytic leukemia. Genes Chromosomes Cancer. 2008;47:781

    793.

    16. Zenz T, Krober A, Scherer K, et al. Monoallelic TP53

    inactivation is associated with poor prognosis in chronic

    lymphocytic leukemia: results from a detailed genetic character-

    ization with long-term follow-up. Blood. 2008;112:33223329.

    17. Rush LJ, Raval A, Funchain P, et al. Epigenetic profiling in

    chronic lymphocytic leukemia reveals novel methylation tar-

    gets. Cancer Res. 2004;64:24242433.

    74 American Society of Hematology

  • 7/30/2019 Leucemias Cronicas Llc1 2010

    6/7

    18. Raval A, Tanner SM, Byrd JC, et al. Downregulation of

    death-associated protein kinase 1 (DAPK1) in chronic lympho-

    cytic leukemia. Cell. 2007;129:879 890.

    19. Kanduri M, Cahill N, Goransson H, et al. Differential genome-

    wide array-based methylation profiles in prognostic subsets of

    chronic lymphocytic leukemia. Blood. 2010;115:296 305.

    20. Fais F, Ghiotto F, Hashimoto S, et al. Chronic lymphocytic

    leukemia B cells express restricted sets of mutated and unmu-

    tated antigen receptors. J Clin Invest. 1998;102:15151525.

    21. Tobin G, Thunberg U, Karlsson K, et al. Subsets with restricted

    immunoglobulin gene rearrangement features indicate a role for

    antigen selection in the development of chronic lymphocytic

    leukemia. Blood. 2004;104:2879 2885.

    22. Messmer BT, Albesiano E, Efremov DG, et al. Multiple distinct

    sets of stereotyped antigen receptors indicate a role for antigen

    in promoting chronic lymphocytic leukemia. J Exp Med.

    2004;200:519525.

    23. Volkheimer AD, Weinberg JB, Beasley BE, et al. Progressive

    immunoglobulin gene mutations in chronic lymphocytic leuke-

    mia: evidence for antigen-driven intraclonal diversification.

    Blood. 2007;109:1559 1567.

    24. Hamblin TJ, Davis Z, Gardiner A, Oscier DG, Stevenson FK.

    Unmutated Ig V(H) genes are associated with a more aggres-sive form of chronic lymphocytic leukemia. Blood. 1999;94:

    18481854.

    25. Rosenwald A, Alizadeh AA, Widhopf G, et al. Relation of gene

    expression phenotype to immunoglobulin mutation genotype in

    B cell chronic lymphocytic leukemia. J Exp Med. 2001;194:

    16391647.

    26. Klein U, Tu Y, Stolovitzky GA, et al. Gene expression profiling

    of B cell chronic lymphocytic leukemia reveals a homogeneous

    phenotype related to memory B cells. J Exp Med. 2001;194:

    16251638.

    27. Seiler T, Woelfle M, Yancopoulos S, et al. Characterization of

    structurally defined epitopes recognized by monoclonal antibod-

    ies produced by chronic lymphocytic leukemia B cells. Blood.

    2009;114:36153624.28. Murray F, Darzentas N, Hadzidimitriou A, et al. Stereotyped

    patterns of somatic hypermutation in subsets of patients with

    chronic lymphocytic leukemia: implications for the role of

    antigen selection in leukemogenesis. Blood. 2008;111:1524

    1533.

    29. Widhopf GF 2nd, Rassenti LZ, Toy TL, Gribben JG, Wierda

    WG, Kipps TJ. Chronic lymphocytic leukemia B cells of more

    than 1% of patients express virtually identical immunoglobu-

    lins. Blood. 2004;104:24992504.

    30. Chu CC, Catera R, Zhang L, et al. Many chronic lymphocytic

    leukemia antibodies recognize apoptotic cells with exposed

    nonmuscle myosin heavy chain IIA: implications for patient

    outcome and cell of origin. Blood. 2010;115:39073915.

    31. Lanham S, Hamblin T, Oscier D, Ibbotson R, Stevenson F,

    Packham G. Differential signaling via surface IgM is associated

    with VH gene mutational status and CD38 expression in

    chronic lymphocytic leukemia. Blood. 2003;101:10871093.

    32. Guarini A, Chiaretti S, Tavolaro S, et al. BCR ligation induced

    by IgM stimulation results in gene expression and functional

    changes only in IgV H unmutated chronic lymphocytic leuke-

    mia (CLL) cells. Blood. 2008;112:782792.

    33. Chen L, Huynh L, Apgar J, et al. ZAP-70 enhances IgM

    signaling independent of its kinase activity in chronic lympho-

    cytic leukemia. Blood. 2008;111:26852692.

    34. Muzio M, Apollonio B, Scielzo C, et al. Constitutive activation

    of distinct BCR-signaling pathways in a subset of CLL patients:

    a molecular signature of anergy. Blood. 2008;112(1):188 195.

    35. Messmer BT, Messmer D, Allen SL, et al. In vivo measure-

    ments document the dynamic cellular kinetics of chronic

    lymphocytic leukemia B cells. J Clin Invest. 2005;115:755

    764.

    36. Calissano C, Damle RN, Hayes G, et al. In vivo intraclonal and

    interclonal kinetic heterogeneity in B-cell chronic lymphocytic

    leukemia. Blood. 2009;114:4832 4842.

    37. Damle RN, Temburni S, Calissano C, et al. CD38 expression

    labels an activated subset within chronic lymphocytic leukemia

    clones enriched in proliferating B cells. Blood. 2007;110:3352

    3359.

    38. Calissano C, Yan XJ, et al. Multi-parameter phenotypic analy-

    sis of members of chronic lymphocytic leukemia clones

    identifies distinct proliferative and resting/re-Entry compart-

    ments with discrete gene expression profiles [abstract 668].

    Blood. 2009;114.

    39. Burger JA, Tsukada N, Burger M, Zvaifler NJ, DellAquila M,

    Kipps TJ. Blood-derived nurse-like cells protect chronic lym-

    phocytic leukemia B cells from spontaneous apoptosis through

    stromal cell-derived factor-1. Blood. 2000;96:26552663.

    40. Burger M, Hartmann T, Krome M, et al. Small peptideinhibitors of the CXCR4 chemokine receptor (CD184) antago-

    nize the activation, migration, and antiapoptotic responses of

    CXCL12 in chronic lymphocytic leukemia B cells. Blood.

    2005;106:18241830.

    41. Nishio M, Endo T, Tsukada N, et al. Nurselike cells express

    BAFF and APRIL, which can promote survival of chronic

    lymphocytic leukemia cells via a paracrine pathway distinct

    from that of SDF-1alpha. Blood. 2005;106:10121020.

    42. Vogler M, Butterworth M, Majid A, et al. Concurrent up-

    regulation of BCL-XL and BCL2A1 induces approximately

    1000-fold resistance to ABT-737 in chronic lymphocytic

    leukemia. Blood. 2009;113:4403 4413.

    43. Kurtova AV, Balakrishnan K, Chen R, et al. Diverse marrow

    stromal cells protect CLL cells from spontaneous and drug-induced apoptosis: development of a reliable and reproducible

    system to assess stromal cell adhesion-mediated drug resis-

    tance. Blood. 2009;114:4441 4450.

    44. Kay NE, Bone ND, Tschumper RC, et al. B-CLL cells are

    capable of synthesis and secretion of both pro- and anti-

    angiogenic molecules. Leukemia. 2002;16:911919.

    45. Ghosh AK, Secreto CR, Knox TR, Ding W, Mukhopadhyay D,

    Kay NE. Circulating microvesicles in B-cell chronic lympho-

    cytic leukemia can stimulate marrow stromal cells: implications

    for disease progression. Blood. 2010;115:17551764.

    46. Muzio M, Scielzo C, Bertilaccio MT, Frenquelli M, Ghia P,

    Caligaris-Cappio F. Expression and function of toll like recep-

    tors in chronic lymphocytic leukaemia cells. Br J Haematol.

    2009;144:507516.

    47. Markey GM, Alexander HD, Agnew AN, et al. Enumeration of

    absolute numbers of T lymphocyte subsets in B-chronic

    lymphocytic leukaemia using an immunoperoxidase technique:

    relation to clinical stage. Br J Haematol. 1986;62:257273.

    48. Goolsby CL, Kuchnio M, Finn WG, Peterson L. Expansions of

    clonal and oligoclonal T cells in B-cell chronic lymphocytic

    leukemia are primarily restricted to the CD3()CD8() T-cell

    population. Cytometry. 2000;42:188 195.

    49. Serrano D, Monteiro J, Allen SL, et al. Clonal expansion within

    the CD4CD57 and CD8CD57 T cell subsets in chronic

    lymphocytic leukemia. J Immunol. 1997;158:14821489.

    Hematology 2010 75

  • 7/30/2019 Leucemias Cronicas Llc1 2010

    7/7

    50. Lotz M, Ranheim E, Kipps TJ. Transforming growth factor beta

    as endogenous growth inhibitor of chronic lymphocytic leuke-

    mia B cells. J Exp Med. 1994;179:999 1004.

    51. Fayad L, Keating MJ, Reuben JM, et al. Interleukin-6 and

    interleukin-10 levels in chronic lymphocytic leukemia: correla-

    tion with phenotypic characteristics and outcome. Blood.

    2001;97:256263.

    52. Beyer M, Kochanek M, Darabi K, et al. Reduced frequencies

    and suppressive function of CD4CD25hi regulatory T cells in

    patients with chronic lymphocytic leukemia after therapy withfludarabine. Blood. 2005;106:2018 2025.

    53. Jak M, Mous R, Remmerswaal EB, et al. Enhanced formation

    and survival of CD4 CD25hi Foxp3 T-cells in chronic

    lymphocytic leukemia. Leuk Lymphoma. 2009;50:788 801.

    54. Gorgun G, Holderried TA, Zahrieh D, Neuberg D, Gribben JG.

    Chronic lymphocytic leukemia cells induce changes in gene

    expression of CD4 and CD8 T cells. J Clin Invest. 2005;115:

    17971805.

    55. Ramsay AG, Johnson AJ, Lee AM, et al. Chronic lymphocytic

    leukemia T cells show impaired immunological synapse forma-

    tion that can be reversed with an immunomodulating drug.J Clin Invest. 2008;118:24272437.

    76 American Society of Hematology


Top Related