You are on page 1of 12

Current Treatment Options in Oncology

DOI 10.1007/s11864-013-0229-5

Leukemia (JP Dutcher, Section Editor)

Update on Chronic
Lymphocytic Leukemia:
Overview of New Agents
and Comparative Analysis
Sanford Kempin, MD
Address
Beth Israel Comprehensive Cancer Center, 325 West 15th Street,
New York, NY 10011, USA
Email: skempin@chpnet.org

* Springer Science+Business Media New York 2013

Keywords Chronic lymphocytic leukemia I New agents I Targeted therapy I Immunomodulation

Opinion statement
Treatment options for lymphoproliferative disorders, including chronic lymphocytic
leukemia (CLL), increasingly are based upon molecular targets, taking advantage of
the immense research output over the past several years elaborating genetic abnormal-
ities, downstream signaling, cell-surface immunobiochemistry, and microenvironmen-
tal stimuli. The latter targets have been particularly useful for the treatment of
multiple myeloma, transforming a previously uniformly, fatal disease to one of a more
chronic and potentially curable disorder. Subsequently, new treatment approaches are
less likely to be based on the more classic types of cytocidal therapy, which, although
successful and essential for the more aggressive disorders that are immediately life-
threatening, tend to be less so, with respect to quality of life, risk versus benefit ratio
and overall curability for the indolent diseases. Because the majority of newer agents
are not available to the clinicians practicing in the community, a number of treatment
options developed over the past two decades are capable of significantly improving the
quality of life of patients with advanced CLL. The initial clinical approach to the patient
should be based on performance status, age, comorbidities, and increasingly on prog-
nostic factors elucidated over the past three decades. Initially, both simple laboratory
studies and easily measurable clinical manifestations were used to guide the clinician
(lymphocyte count, anemia, thrombocytopenia, enlarging lymph nodes, splenomegaly,
hepatomegaly), and clinical staging systems were developed. At present a cadre of bi-
ologic factors, including cytogenetic alterations, gene expression profiles with subse-
quent immunoglobulin abnormalities, and expression of CD38 and Zap-70, are now
available and are standard decision-making criteria to treat a patient with CLL. An ini-
tial period of observation allows the clinician along with the patient to gather all the
information necessary to make an informed treatment decision. Frequently, a “watch
and wait” approach, which for CLL does not appear to harm the patient, is the most
Leukemia (JP Dutcher, Section Editor)

appropriate decision. Complications of CLL, such as autoimmune hemolytic anemia and


idiopathic thrombocytopenic purpura, will lead to treatment at least temporarily in
those patients who might otherwise have not needed therapy. Frontline therapy will
range from easily administrable single-agents to combination chemoimmunotherapy
regimens. Experimental protocols, utilizing “post state of the art” treatments, are
available in the form of research protocols at major treatment centers. At the present
time, it is premature to recommend bone marrow ablative therapy as initial treatment
unless the prognosis appears grave and the patient can withstand the rigors of
this approach.

Introduction
CLL is the most commonly encountered leukemia, of the disease. With the application of the Rai clinical
with an incidence in the United States of 22–30 per staging system [3], one can begin to appreciate the
100,000 in patients older than age 65 years and a me- benefit, or lack thereof, of these therapeutic interven-
dian age at diagnosis of 72 years [1]. At present, both tions. During the past two decades, an increasing
internists and hematologists are observing that “rou- number of prognostic factors not only have opened
tine” physical evaluations in younger patients often the door to understanding the clinical heterogeneity
demonstrates abnormalities of the complete blood of the disease but have led to possible therapeutic
count, particularly the neutrophil/lymphocyte ratio, implications not previously appreciated. As we have
which lead to further evaluations and a diagnosis of begun to understand the natural history of CLL, the
a clonal B-cell proliferation.. The clinical manifesta- need for observation versus treatment has become
tions of CLL range from an asymptomatic patient with the most important determinant in a clinician’s initial
minimal B-cell lymphocytosis to a progressive clinical approach to the patient.
picture of enlarging lymph nodes, splenomegaly, ane- Recognized since the modern era of treatment of
mia, thrombocytopenia, and life-threatening infections. the disease were patients who developed a lymphocy-
Historically, the treatment for CLL has been one of tosis consistent with CLL but in whom treatment was
an eradicative approach utilizing agents and proce- unnecessary and survival was similar to an aged-
dures with direct cell-kill capacity. Radiation therapy matched control population. Benign monoclonal lym-
was the initial therapy of CLL and numerous treatment phocytosis (BML) [4] has been studied for the past de-
modalities were explored, some of which were quite cade, and although this entity itself may have its own
successful in controlling the disease and normalizing heterogeneity, the basic mechanisms responsible for
blood counts [2]. Subsequently, single- and multiple- its lack of progression in most patients into a symp-
agent chemotherapy and more recently combinations tomatic disorder have not been elucidated. The impor-
of chemotherapy and monoclonal antibodies have be- tance of recognizing this population is to prevent the
come the standard approaches. With our increasing utilization of unnecessary and even potentially lethal
ability to understand, at a molecular level, the basic treatment in the absence of clinical indications.
properties of B cells, such as differentiation, prolifera- It is essential that a period of observation be under-
tion, cell death-apoptosis, and crosstalk between other taken before a treatment decision is made unless life-
lymphoid and nonlymphoid cell populations in the threatening complications are present. Once clinical
environmental niches, it becomes evident that there acumen and laboratory parameters lead to a decision
are now an increasing number of targets, leading to to treat the patient, a variety of options are available.
what might now be called “downstream therapeutics,” Risk stratification is helpful when deciding upon a
taking advantage of the structure and function of treatment modality [5••]. These options, depending
these targets. upon the clinical situation, may include the use of
During the past 100 years, numerous therapeutic alkylating agents alone to the more recent combina-
trials have been reported for the treatment of CLL. tion therapies developed over the past two decades
These trials have been used with less than an ideal [6, 7, 8••, 9]. Recently, a number of new agents have
appreciation of the heterogeneity and natural history demonstrated important clinical efficacy, and some
Update on Chronic Lymphocytic Leukemia: Overview of New Agents Kempin

have been approved for use in CLL, but it is not yet to cure the disease, this accumulation of new agents
possible to compare their overall efficacy to the more offers the future potential for an” almost” cure (i.e., ty-
standard regimens. However, in view of the fact that rosine kinase inhibitors for the treatment of chronic
less than marrow ablative approaches are not likely myelogenous leukemia) with less morbidity.

Current Treatment Approaches in the Modern Era


Before the introduction of purine nucleoside analogues (PNA), the concept
of cure of CLL was not seriously considered as a therapeutic goal except at
centers devoted to stem cell transplantation. Alkylating agents were success-
ful at controlling clinical manifestations and normalizing blood counts.
However, these treatments appeared to have only modest impact on survival.
Patients inexorably relapsed from what were only partial remissions. Com-
bination chemotherapy, beginning with the introduction of corticosteroids
[10], was developed both in the United States and Europe [11–13], and al-
though significant numbers of complete responders were reported in various
trials, there was no evidence that eradication of the disease had occurred and
patients eventually relapsed. It was clear that to eradicate this disease one had to
be able to demonstrate the absence of minimal residual disease (MRD) if one
were to hope for eradication of the malignant clone(s) of cells and restoration of
normal hematopoiesis [14]. Before the modern era of therapy, this remained an
elusive endpoint, other than in the setting of marrow ablative approaches
[15••]. PNA therapy was introduced in the early 1970s and in combination with
alkylating agents and monoclonal antibodies has become the standard treat-
ment approach. Responses to these combinations in appropriately chosen pa-
tient populations are successful in achieving long-term, disease-free intervals as
well as a survival advantage compared with earlier treatments.

Purine Nucleoside Analogues (Pentostatin, Cladribine, Fludarabine)


Deoxycoformycin (Pentostatin (P); Bedford) was the first purine analogue to
enter clinical trials, and the results as reported by Grever et al. were impressive
[16]. By combining it with an alkylating agent (cyclophosphamide) and a CD-
20 monoclonal antibody (PCR), a particularly effective combination is now in
use as front-line therapy [17, 18]. Cladribine (Leustatin; Janssen Biotech)
demonstrated promising results with complete and partial remissions of 38 %
and 37 % respectively in untreated patients [19]. Grade III and IV toxicities were
significant, and this agent is only rarely used today for this indication.
Fludarabine (Fludara; Genzyme) was initially developed for the treatment of
acute myeloid leukemia. However, at marrow ablative doses this agent was le-
thal. By pharmacokinetic manipulation, Keating et al. [20] were able to establish
a therapeutic dose that was highly effective at inducing remissions. The addition
of an alkylating agent (cyclophosphamide) [21] and a CD-20 monoclonal an-
tibody-rituximab (FCR) [22] has given similar responses to the PCR combina-
tion, and both are now considered frontline therapies. It should not, however,
be forgotten that alkylating agents, as a single modality, in patients in whom
disease “eradication” would appear to be unrealistic, is still a reasonable ap-
proach [23]. Unfortunately, even with such aggressive therapy (PCR, FCR), the
Leukemia (JP Dutcher, Section Editor)

eradication of the disease, at least at the molecular level, is still not possible and
short- and long-term side effects, such as immunosuppression with resultant
infections [24••] and second malignancies (a complication known to be in-
creased in patients with CLL as a reflection of its natural history) have been
reported [25]. The encumbrance of administration and toxicities of these agents
in combination, however, is likely to decrease, if not abolish their use, as more
directed approaches, taking advantage of the pathophysiology of CLL cells and
their environmental niche, are found to be at least as equally effective and less
toxic. Bendamustine (Treanda; Cephalon) was produced to take advantage of
both the alkylating agent and purine nucleoside properties of a single molecule
[26]. At present, the drug is thought to act predominantly as an alkylating agent.
Both as a single agent and in combination with monoclonal antibodies,
bendamustine has demonstrated significant activity in non-Hodgkin lympho-
ma and CLL [27] and currently is being used as initial therapy and in numerous
clinical trials.

Stem-Cell Transplantation
Stem-cell cell transplantation has been utilized for nearly three decades for
the treatment of CLL and the most recent updates confirm its efficacy in
eradicating the disease [28••]. Previously limited to younger patients with
minimal comorbidities, clinical situations that are unusual in a standard CLL
population, reduced intensity conditioning offers older patients an oppor-
tunity for this approach [28••]. Adoptive immunity utilizing modified T cells
to recognize B-cell antigens is the most intriguing of new therapies and may
ultimately replace our standard treatment regimens [29••].

Monoclonal Antibody Therapy


An ideal therapeutic intervention would consist of targeting a specific molecule,
one found only on the malignant cell. Studies on the immunoglobulin-ex-
pressing B cell demonstrated the feasibility of this approach in non-Hodgkin
lymphoma, and in fact long-term survival was demonstrated [30]. This ap-
proach utilizing unique idiotypes to produce the antibodies, being labor in-
tensive, led investigators to direct efforts toward the production of antibodies
utilizing less specific but more generalized tumor antigens as targets. In CLL,
mainly unconjugated antibodies have been used. Examples of such targets in-
clude CD 20 [31] and CD 52 [32]. The mechanisms of tumor cell destruction
include antibody-dependent cell cytotoxicity (ADCC), complement-dependant
cytotoxicity (CDC), and signaling pathways of apoptosis.
Rituximab (Rituxan; Genentech), the first monoclonal antibody approved
for the treatment of lymphoproliferative disorders, is directed against the CD
20 epitope, important in calcium transport. Since Phase I studies were
completed, there have been numerous reports of its efficacy as a single agent
in CLL [33]. The results were modest compared with its use in non-Hodgkin
lymphomas. Decreased expression of CD 20 was thought to be the pre-
dominant reason for this difference [34]. Modulation of surface CD20 by
bortezomib (Velcade; Millennium), epigenetic alteration of gene expression
and increase in dose may improve its efficacy [35–37]. A significant deter-
minant of its efficacy is related to single nucleotide polymorphisms of
FcgRIIIa and FcgRIIa [38]. The use of rituximab has now become part of the
Update on Chronic Lymphocytic Leukemia: Overview of New Agents Kempin

standard front-line regimen in CLL after studies showing significant activity


when combined with a purine nucleoside analogue. The German CLL Study
group demonstrated significant benefit compared with fludarabine alone
[39]. The triplet utilizing FCR was more successful than FC [40], thus
establishing the rational for triplet therapy in the frontline or relapse
setting. Rituximab has been found to be useful in treating several com-
plications of CLL (and other lymphomas), including idiopathic throm-
bocytopenic purpura (ITP) [41], autoimmune hemolytic anemia (AIHA)
[42], and red cell aplasia (RCA) [43]. Rituximab maintenance therapy after
maximal initial response has become standard practice for the treatment
of non-Hodgkin lymphoma and has been explored for the treatment of
CLL [44].

Campath-1H
Campath-1H (Alemtuzumab; Genzyme), a monoclonal antibody, is directed
against the CD52 antigen, expressed on lymphocytes, at nearly all stages of
differentiation, as well as granulocytes, eosinophils, monocytes, and mac-
rophages. Significant activity has been well documented [45]. Its mechanism
of action is related to ADCC, CDC, and direct cell death and appears to be
independent of the p53 abnormality noted in resistant CLL [46]. Subcuta-
neous administration has led to a more easily administrable regimen [47].
This represents a reasonable single-agent to treat refractory patients.
Antibacterial and antiviral prophylaxis are used during its administration as
infectious complications, particularly reactivation of CMV may be life-
threatening in this population [24••]. Alemtuzumab has been used along
with a PNA combination [48]. When used as a consolidation program after
maximal efficacy of one of the PNA regimes, significant toxicity was observed
[49, 50]. Consolidation after PCR is still being explored as part of a clinical
trial. Its ability to improve blood and bone marrow involvement has led to
its use as a preparatory regimen for stem-cell transplantation [51].

Ofatumumab (Arzerra; GlaxoSmithKline and Genmab A/S)


This agent, specific for the CD20 molecule but distinct from the epitope
recognized by rituximab, is reported to have more CDC activity than
rituximab [52]. In combination with fludarabine and cyclophosphamide,
it has significant activity in the initial treatment of high-risk patients
[53]. However, its overall efficacy, particularly in patients with the 17p
deletion and bulky disease, is modest.

Other Monoclonal Antibodies and Immunoconjugates


The CD20 molecule is the target for GA101 (obinutuzumab) a type II hu-
manized monoclonal antibody, which differs in its binding properties from
the type I agents, such as rituximab and ofatumumab [54]. The benefits,
relative to rituximab, include lack of internalization as seen with type I agents
and induction of nonapoptotic cell death through lysosomal mediation [55].
A number of monoclonal antibodies are in clinical development, includ-
ing HU1D10, a humanized monoclonal antibody, developed against a var-
iant of an epitope on the HLA-DR B chain [56]. Clinical studies have
demonstrated activity in CLL [57]. Lumiliximab, directed against CD 23, has
Leukemia (JP Dutcher, Section Editor)

demonstrated activity for inducing ADCC and apoptosis in CLL cells and clinical
trials, particularly in combination with FCR, have been reported recently, with
disappointing results [58]. TRU-016, a modification of a CD 19 antibody has
completed phase 1 clinical trials [59]. Other antibodies being explored include
those targeting CD 19 (XmAb5574) and CD 22 (epratuzumab) [60].
It is unclear at this time whether further attempts at using monoclonal an-
tibodies, as single-treatment modalities, will be undertaken. Rather, their
combinations with other specific targeted therapies may yield significant
advances. Immunoconjugates represent an attempt to improve the thera-
peutic efficacy of monoclonal antibodies. LMB −2, a conjugate utilizing a
pseudomonas exotoxin, directed against CD25 demonstrated significant re-
sponses in CLL [61] as has the anti CD-22 immunotoxin BL-22 [62]. It is not
known whether further immunoconjugates will be developed specifically for
CLL.

Innovative Biologic Therapy


Recent insights into the biology of CLL cells and their interaction with the
microenvironment (mesenchymal stromal cells, monocyte-derived nurse-like
cells, T cells, and matrix) have led to new targets for agents that may be used
singly or added to more standard regimens [63, 64, 65••]. Bone marrow
stromal cells (BMSC) provide both the structural integrity to support CLL cell
growth as well as growth factors. There is a bidirectional cross-link between
BMSCs and CLL cells; the relationship is maintained by chemokine receptors
(CXCR4 and CXCR5) and adhesion molecules (VLA-4). The B-cell receptor
(BCR) is essential to maintain the B-cell clonal expansion through down-
stream interactions with a series of tyrosine kinases, including spleen tyrosine
kinase (Syk) and Bruton tyrosine kinase (BTK) and secretion of cytokines
(CCL3) functioning as chemoattractants for T cells and monocytes [63, 64].

B-Cell Receptor Signaling Pathway and Kinase Inhibitors


Dasatinib (Sprycel; Bristol-Myers Squibb): This broad-spectrum tyrosine
kinase inhibitor, presently used as up-front therapy of chronic myelog-
enous leukemia, demonstrates modest activity in CLL with
myelosuppression as the predominant side-effect [65••, 66••, 67, 68].
Ibrutinib (PCI-32765): This is an irreversible inhibitor of BTK [69••].
This agent also has been found to inhibit microenvironmental stimuli
[70••]. Significant activity has been observed in CLL with an acceptable
side effect profile. Lymphocytosis is a common occurrence in this class
of agents. Studies are ongoing, combining ibrutinib with monoclonal anti-
bodies and bendamustine [71••]. Further combination studies are anticipated.
Fostamatinib (R406 pro-drug): This is a reversible inhibitor of Syk. Modest
activity has been observed in SCLL/CLL with minimal toxicity. Lymphocytosis is
commonly observed [72].
GS-101 (CAL-101): This is a reversible inhibitor of P13KD form of the
p110 catalytic subunit [73]. As a part of the BCR activation sequence,
P13K appears to be constitutively active in CLL. Inhibitors of this group
promote caspase-dependent apoptosis and inhibit the protective nature
of stromal cell (nurse cells) interaction with CLL cells, with downregulation of
chemokines and cytokines. The major side-effects are liver function ab-
Update on Chronic Lymphocytic Leukemia: Overview of New Agents Kempin

normalities and, as expected, lymphocytosis. This agent has been com-


bined with rituximab and bendamustine with an acceptable side effect
profile [74].

BCL-2 Inhibition
Oblimersen sodium: BCL-2 is universally expressed and upregulated in CLL
cells, possibly related to a deletion of miRNA regulators. Antisense oli-
gonucleotides that bind mRNA, inducing enzymatic cleavage and inhibi-
tion of protein translation, have been developed during the past 20 years
and have been utilized in BCL-2–expressing lymphoid malignancies. The
first to enter clinical trials in CLL was oblimersen sodium, which dem-
onstrated modest activity as a single agent [75]. Myelosuppression was the
major toxicity. First cycle reactions, such as tumor-lysis syndrome, were
reported. This agent has been combined with rituximab and cyclophos-
phamide, demonstrating an increase in the CR/nPR rate [76]. There are
no plans at this time for further development.
ABT-263 (Navitoclax): This agent inhibits BCL-2, BCL-xl, and BCL-w, induc-
ing apoptosis of CLL cells [77]. Responses were seen in fludarabine refractory
patients and this drug is now in clinical trials [78]. Agents in this group of drugs
might sensitize the cells to the apoptotic effects of rituximab [79].
Homoharringtonine: This inhibitor of Mcl-1, a member of the Bcl-2 fam-
ily of antiapoptotic proteins, was developed originally for the treatment of
acute myeloid leukemia and now used for the treatment of CML, has dem-
onstrated significant in vitro activity against CLL cells and will be entering
into clinical trials [80].

Immunomodulation
Lenalidomide (Revlimid; Celgene): The introduction of immunomodulating
drugs (IMiDs) has revolutionized the treatment of multiple myeloma [81]
and improved patients with the myelodysplastic syndrome [82].
Lenalidomide, a thalidomide analogue, apart from its significant activity in
MDS and multiple myeloma, has been shown to decrease the production of
growth factors, which prolong the survival of CLL cells [83]. These include
vascular endothelial growth factor and TNF-a, which may alter the micro-
environment in favor of CLL proliferation. The overall response rate in re-
lapsed or refractory CLL patients was significant, but with no evidence of
specificity for the 17p resistant patients [84••]. Life-threatening tumor flares
and a tumor lysis syndrome were observed at a dose of 25 mg/d for 21 days
[85] and safer schedules are being explored. It is feasible to combine
lenalidomide and monoclonal antibodies [86, 87] and chemotherapy [88];
however, significant toxicity remains a problem and the most appropriate
treatment schedule for lenalidomide has not yet been determined.

Conclusions
The treatment of the symptomatic patient with CLL remains a therapeutic
challenge. The availability of active agents and combinations has improved
the quality of life and for some has resulted in a survival advantage. How-
ever, the “cure” for the disease, defined as best as we can by ablation of clonal
Leukemia (JP Dutcher, Section Editor)

lymphopoiesis, and normalization of the immune response has not yet been
accomplished. The initiating event in this disease is still mysterious; the
primeval immune dysregulatory event has not been defined, taking into
consideration the numerous genetic abnormalities discovered. The familial
nature of immune disorders seen in this population and its association with
aging favor a still undefined genomic basis. Because normalization of the
immune system is still not possible other than by allogeneic reconstitution,
our present strategies will consist of treating those manifestations of the
disease as described and at the same time exploring new therapeutic strate-
gies. With clinical expertise, the improvement in quality of life of patients
with this disorder should be the most important goal. As reviewed, at least a
number of options are available to patients, and over the next several years
many of the new treatments being explored are likely to become standard.
However, unless combined with basic research programs, the disease is not
likely to be eradicated. The most recent studies of clonal evolution of CLL [89,
90] demonstrate the need for early initial, radical elimination of the malignant
cell before resistance may occur or the need for multiple targeted therapy—ei-
ther upfront or sequentially—an approach that has been highly successful for
the treatment of AIDS.

Conflict of Interest
Sanford Kempin is a consultant for Celgene.

References and Recommended Reading


Papers of particular interest, published recently, have been
highlighted as:
•• Of major importance

1. Ries LAG, Melbert D, Krapcho, et al. SEER Clinical chronic lymphocytic leukemia. J Clin Oncol.
Statistics Review, 1975–2005. Bethesda: National 2009;27:4378–84.
Cancer Institute; 2008. 7. Woyach JA, Ruppert AS, Heerema NA, et al.
2. Kempin S, Shank B: Radiation in chronic lymphocytic Chemoimmunotherapy with fludarabine and
leukemia in Chronic Lymphocytic Leukemia :Recent rituximab produces extended overall survival and
Progress and Future Directions. Alan R. Liss; 1987. 337- progression – free survival in chronic lymphocytic
352 leukemia: long-term follow-up of CALGB study
3. Rai KR, Sawitsky A, Cronkite EP, et al. Clinical 9712. J Clin Oncol. 2011;29:1349–55.
staging if chronic lymphocytic leukemia. Blood. 8.•• Badoux XC, Keating MJ, Wang X, et al. Fludarabine,
1975;46:219–34. cyclophosphamide, and rituximab
4. Ghia P, Caligaris-Cappio F. Monoclonal B-cell chemoimmunotherapy is highly effective treatment
lymphocytosis: right track or red herring. Blood. for relapsed patients with CLL. Blood.
2012;119:4358–62. 2011;117:3016–24.
5.•• Gribben JG, O’Brien S. Update on therapy of This is the most important long-term follow-up manuscript
chronic lymphocytic leukemia. J Clin Oncol. describing the results of the most frequently used treatment
2011;29:544–50. of CLL.
An excellent review of current therapeutic options, intro- 9. Lamanna N, Kalaycio M, Maslak P, et al. Pentostatin,
ducing as well newer experimental agents. cyclophosphamide, and rituximab is an active,
6. Knauf WU, Lissichkov T, Aldaoud A, et al. Phase III well-tolerated regimen for patients with previously
randomized study of Bendamustine compared with treated chronic lymphocytic leukemia. J Clin Oncol.
chlorambucil in previously untreated patients with 2006;24:1575–81.
Update on Chronic Lymphocytic Leukemia: Overview of New Agents Kempin

10. Han T, Ezdinli EZ, Stutzman L. Chlorambucil vs. 21. Eichhorst BF, Busch R, Hopfinger G, et al.
combined chlorambucil corticosteroid therapy in Fludarabine plus cyclophophamide versus
chronic lymphocytic leukemia. Cancer. fludarabine alone in first-line therapy of younger
1973;31:502–8. patients with chronic lymphocytic leukemia. Blood.
11. Kempin SJ, Lee BJ, Thaler HT, et al. Combination 2006;107:885–9.
chemotherapy of advanced chronic lymphocytic 22. Tam CS, O’Brien S, Wierda W, et al. Long-term results
leukemia: the M-2 protocol (vincristine, BCNU, of the fludarabine, cyclophosphamide, and
cyclophosphamide, melphalan, and prednisone). rituximab regimen as initial therapy of chronic
Blood. 1982;60:1110–21. lymphocytic leukemia. Blood. 2008;112:975–80.
12. French Cooperative Group on Chronic Lymphocytic 23. Eichhorst BF, Busch R, Stilgenbauer S, et al. First-line
Leukemia. Long –term results of the CHOP regimen therapy with fludarabine compared with
in stage C chronic lymphocytic leukaemia. Br J chlorambucil does not result in a major benefit for
Haematol. 1989;73:334–40. elderly patients with advance chronic lymphocytic
13. Oken MM, Kaplan ME. Combination chemotherapy leukemia. Blood. 2009;114:3382–91.
with cyclophosphamide, vincristine, and prednisone 24.•• Elter T, Vehreschild JJ, Gribben J, et al. Management
in the treatment of refractory chronic lymphocytic of infections in patients with chronic lymphocytic
leukemia. Cancer Treat Rep. 1979;63:441–7. leukemia treated with alemtuzumab. Ann Hematol.
14. Rawstron C, Kennedy B, Evans PA, et al. Quanti- 2009;88:121–32.
tation of minimal residual disease in chronic This excellent review of an important subject which must be
lymphocytic leukemia using a sensitive flow taken into consideration in any planned therapy of CLL.
cytometric assay improves the prediction of out- 25. Cheson BD, Vena DA, Barrett J, Freidlin B. Second
come and can be used to optimize therapy. Blood. malignancies as a consequence of nucleoside analog
2001;98:29–35. therapy for chronic lymphoid leukemias. J Clin
15.•• Dreger P, Dohner H, Ritgen M, et al. Allogeneic Oncol. 1999;17:2454–2460.
stem-cell transplantation provides durable disease 26. Leoni LM, Bailey B, Reifert J, et al. Bendamustine
control in poor-risk chronic lymphocytic leukemia (Treanda) displays a distinct pattern of cytotoxicity
(CLL): long-term clinical and MRD results of the and unique mechanistic features compared with
German CLL study group CLL3X trial. Blood. other alkylating agents. Clin Cancer Res.
2010;16:2438–47. 2008;14:309–17.
An important study allowing investigators to compare the
most radical treatment of CLL (stem-cell transplantation) 27. Bergmann MA, Goebeler MA, Herold M, et al.
with conventional therapy. Although not a randomized Efficacy of bendamustine in patients with relapsed or
study there is a great deal of useful information. refractory chronic lymphocytic leukemia: results of a
phase I/II study of the German CLL Study Group.
16. Grever MR, Siaw MP, Jacob WF, et al. The biochem- Haematologica. 2005;90:1357–64.
ical and clinical consequences of 2’-deoxycoformycin
in refractory lymphoproliferative malignancy. Blood. 28.•• Kharfan-Dabaja MA, Bazarbachi A. Hematopoietic
1981;57:406–17. stem cell allografting for chronic lymphocytic
17. Weiss MA, Maslak P, Jurcic JG, et al. Pentostatin and leukemia: a focus on reduced-intensity conditioning
cyclophosphamide: A new effective regimen in regimens. Cancer Control. 2012;19:68–75.
previously treated patients with chronic lymphocytic This manuscript represents an important advance for the
leukemia (CLL). J Clin Oncol. 2003;21:1278–84. potential eradication of CLL in an older population.
18. Kay NE, Geyer SM, Call TG, et al. Combination 29.•• Kochenderfer JN, Wilson WH, Janik JE, et al.
chemoimmunotherapy with pentostatin, cyclophos- Eradication of B-lineage cells and regression of
phamide, and rituximab shows significant clinical lymphoma in a patient treated with autologous T
activity with low accompanying toxicity in previously cells genetically engineered to recognize CD19.
untreated B chronic lymphocytic leukemia. Blood. Blood. 2010;116:4099–102.
2007;109:405–11. This manuscript represents a new approach now being un-
19. Juliusson G, Liliemark J. High complete remission dertaken by a number of investigators which may radically
rate from 2-chloro-2’deoxyadenosine in previously change our approach to treatment.
treated patients with B-cell chronic lymphocytic 30. Vose J. Personalized immunotherapy for the treatment
leukemia: response predicted by rapid decrease of of non-Hodgkin lymphoma: a promising approach.
blood lymphocyte count. J Clin Oncol. Clin Advan Hematol Oncol. 2005;3:923–32.
1993;11:679–89. 31. McLaughlin P, Grillo-Lopez AJ, Link BK, et al.
20. Keating MJ, O’Brien S, Lerner S, et al. Long term Rituximab chimeric anti-CD-20 monoclonal
follow-up of patients with chronic lymphocytic antibody therapy for relapsed indolent lymphoma.
leukemia (CLL) receiving fludarabine regimens as Half of patients respond to a four-dose treatment
initial therapy. Blood. 1998;92:1165–71. program. J Clin Oncol. 1998;16:2825–33.
Leukemia (JP Dutcher, Section Editor)

32. Osterborg A, Dyer M, Bunjes D, et al. Phase II with B-cell chronic lymphocytic leukemia. Cancer.
multicenter study of human CD52 antibody in 2008;112:119–28.
previously treated chronic lymphocytic leukemia. J 45. Keating MJ, Flinn I, Jain V, et al. Therapeutic role of
Clin Oncol. 1997;15:1567–74. alemtuzumab (Campath-1H) in patients who have
33. Hainsworth JD, Litchy S, Barton JH, et al. Single- failed fludarabine: Results of a large international
agent rituximab as first-line and maintenance study. Blood. 2002;99:3554–61.
treatment for patients with chronic lymphocytic 46. Stilgenbauer S, Dohner H. Campath-1H-induced
leukemia or small cell lymphocytic lymphoma: a complete remission of chronic lymphocytic leukemia
phase II trial of the Minnie Pearl Cancer Research despite p53gene mutation and resistance to
Network. J Clin Oncol. 2003;212:1746–51. chemotherapy. N Engl J Med. 2002;347:452–3.
34. Manshouri T, Do KA, Wang X, et al. Circulating 47. Lundin J, Kimby E, Bjorkholm M, et al. Phase II trial
CD20 is detectable in the plasma of patients with of subcutaneous antiCD52 monoclonal antibody
chronic lymphocytic leukemia and is of prognostic alemtuzumab (Campath-1H) as first-line treatment
significance. Blood. 2003;101:2507–13. for patients with B-cell chronic lymphocytic
35. Bill J, Winiarska M, Nowis D, et al. Bortezomib leukemia (B-CLL). Blood. 2002;100:768–73.
modulates surface CD20 in B-cell malignancies and 48. Badoux XC, Keating MJ, Wang X, et al. Cyclophos-
effects rituximab-mediated complement-dependent phamide, fludarabine, alemtuzumab, and rituximab
cytotoxicity. Blood. 2010;115:3745–55. as salvage therapy for heavily pretreated patients with
36. Hiraga J, Tomita A, Sugimoto T, et al. Down-regulation chronic lymphocytic leukemia. Blood.
of CD20 expression in B-cell lymphoma cells after 2011;118:2085–93.
treatment with rituximab-containing combination 49. Lepetre S, Aurran, T, Mahé B, et al. Excess mortality after
chemotherapies: its prevalence and clinical signifi- treatment with fludarabine and cyclophosphamide in
cance. Blood. 2009;113:4885–93. combination with alemtuzumab in previously untreated
37. O’Brien S, Kantarjian H, Thomas DA, et al. Rituximab patients with chronic lymphocytic leukemia in a ran-
dose-escalation trial in chronic lymphocytic leukemia. J domized phase 3 trial. Blood. 2012;119:5104–10.
Clin Oncol. 2001;19:2153–64. 50. Lin TS, Donohue KA, Byrd JC, et al. Consolidation ther-
38. Cartron G, Dacheux L, Salles G, et al. Therapeutic apy with subcutaneous alemtuzumab after fludarabine
activity of humanized anti CD20 monoclonal and rituximab induction therapy for previously
antibody and polymorphism in IgG Fc receptor untreated chronic lymphocytic leukemia: final analysis
FcgammaRIIIa gene. Blood. 2002;99:754–8. of CALGB 10101. J Clin Oncol. 2010;28:4500–6.
39. Woyach JA, Ruppert AS, Heerema NA, et al. 51. Delgado J, Thomson K, Russell N, et al. Results of
Chemoimmunotherapy with fludarabine and alemtuzumab based reduced-intensity allogeneic
rituximab produces extended overall survival and transplantation for chronic lymphocytic leukemia: a
progression-free survival in chronic lymphocytic British Society of Blood and Marrow Transplantation
leukemia: Long- term follow-up of CALGB Study Study. Blood. 2006;107:1724–30.
9712. J Clin Oncol. 2011. 52. Pawluczkowycz A, Beurskens F, Beum P, et al. Binding
40. Keating MJ, O’Brien S, Albitar M, et al. Early results of of submaximal C1q promotes complement dependent
a chemoimmunotherapy regimen of fludarabine, cytotoxicity (CDC) of B cells opsonized with anti-
cyclophosphamide, and rituximab as initial therapy CD20 mAbs ofatumumab (OFA) or rituximab (RTX):
for chronic lymphocytic leukemia. J Clin Oncol. considerably higher levels of CDC are induced by OFA
2005;23:4079–88. than by RTX. J Immunol. 2009;183:749–58.
41. Hegde UP, Wilson WH, White T, Cheson BD. 53. Wierda W, Kipps TJ, Durig J, et al.
Rituximab treatment of refractory fludarabine Chemoimmunotherapy with O-FC in previously
associated immune thrombocytopenia in chronic untreated patients with chronic lymphocytic
lymphocytic leukemia. Blood. 2002;100:2260–2. leukemia. Blood. 2011;117:6450–8.
42. Shvidel L, Shalrid M, Berrebi A. Intractable 54. Niederfellner GJ, Lammens A, Schwaiger M, et al. Crys-
autoimmune hemolytic anemia in B cell chronic tal structure analysis reveals that the novel type II anti-
lymphocytic leukemia resolved by rituximab. Leuk CD20 antibody GA 101 interacts with a similar epitope
Lymphoma. 2004;45:1493–4. as rituximab and ocrelizumab but in a fundamentally
43. Narra K, Borghael H, Al-Saleem T, Hoglund M, Smith different way. Blood. 2009;114:3726. abstract.
MR. Pure-red cell aplasia in B-cell 55. Alduaij W, Ivanov A, Honeychurch J, et al. Novel type
lymphoproliferative disorder treated with rituximab: II anti-CD20 monoclonal antibody (GA 101) evokes
report of two cases and review of the literature. Leuk homotypic adhesion and actin-dependent, lysosome-
Res. 2006;30:109–14. mediated cell death in B-cell malignancies. Blood.
44. Del Poeta G, Del Principe MI, Buccisano F, et al. 2011;117:4519–29.
Consolidation and maintenance immunotherapy 56. Brown KS, Levitt DJ, Shannon M, Link BK. Phase II
with rituximab improve clinical outcome in patients trial of Remitogen (humanized 1D10 monoclonal
Update on Chronic Lymphocytic Leukemia: Overview of New Agents Kempin

antibody targeting class II in patients with relapsed receptor-and chemokine-controlled adhesion and
low-grade or follicular lymphoma. Clin Lymphoma. migration in chronic lymphocytic leukemia. Blood.
2001;2:188–90. 2012;119:2590–4.
57. Byrd JC, Lucas M, Cheson BD, Flinn IW. Monoclonal This important manuscript documents the rationale for
antibody therapies for patients with chronic treatment with this agent.
lymphocytic leukemia. In: Cheson BD, editor. 70.•• Herman SE, Gordon AL, Hertlein E, et al. Bruton
Monoclonal Antibody Therapy of Hematologic tyrosine kinase represents a promising therapeutic
Malignancies. Abingdon: Darwin Scientific target for treatment of chronic lymphocytic leukemia
Publishing Limited; 2001. p. 91–112.
and is effectively targeted by PCI-32765. Blood.
58. Byrd JC, Kipps TJ, Flinn IW, et al. Phase1/2 study
2011;117:6287–96.
of lumiliximab combined with fludarabine,
This study confirms the rationale for targeted therapy with a
cyclophosphamide, and rituximab in patients with
new agent likely to become part of our future therapy.
relapsed or refractory chronic lymphocytic leukemia.
Blood. 2010;115:489–95. 71.•• O’Brien SM, Barrientos JC, Flinn IW, et al. Combi-
nation of the Bruton’s tyrosine kinase (BTK) inhibi-
59. Awan FT, Lapalombella R, Trotta R, et al. CD19
tor PCI-32765 with bendamustine (B)/rituximab
targeting of chronic lymphocytic leukemia with a
(R)(BR) in patients(pts) with relapsed/refractory
novel Fc-domain-engineered monoclonal antibody.
(R/R) chronic lymphocytic leukemia (CLL): Interim
Blood. 2010;115:1204–13.
results of a phase Ib/II study. J Clin Oncol.
60. Leonard JP, Coleman M, Ketas JC, et al. Phase I/II 2012;30:6515
trial of epratuzumab (humanized anti CD22 These results will form the basis of planned clinically trials
antibody) in indolent non-Hodgkin’s lymphoma. J utilizing this new agent in randomized studies.
Clin Oncol. 2003;21:3051–9.
72. Friedberg JW, Sharman J, Sweetenham J, et al.
61. Kreitman RJ, Wilson WH, White JD, et al. Phase Inhibition of Syk with fostamatinib disodium
I trial of recombinant immunotoxin Anti-Tac has significant clinical activity in non-Hodgkin
(Fv)-PE38(LMB-2) in patients with hematologic lymphoma and chronic lymphocytic leukemia.
malignancies. J Clin Oncol. 2000;18:1614–36. Blood. 2010;115:2578–85.
62. Kreitman RJ, Pastan I. Antibody fusion proteins: anti- 73. Lannutti BJ, Meadows SA, Herman SEM, et al. CAL-
CD22 recombinant immunotoxin moxetumomab 101, a p110d selective phosphatidylinositol-3-kinase
pasudotox. Clin Cancer Res. 2011;17:6398–405. inhibitor for the treatment of B-cell malignancies,
63. Woodland RT, Fox CJ, Schmidt MR, et al. Multiple inhibits P13K signaling and cellular viability. Blood.
signaling pathways promote B lymphocyte stimula- 2011;117:591–4.
tor dependent B-cell growth and survival. Blood. 74. Flinn IW, Schreeder MT, Coutre SE, et al. A phase 1
2008;111:750–60. study of CAL-101, an isoform-selective inhibitor of
64. Burger JA. Nurture versus Nature: the microenviron- phosphatidylinositol 3-kinase P110 (delta), in
ment in chronic lymphocytic leukemia. Hematology combination with anti-CD20 monoclonal antibody
Am Soc Hematol Educ Program. 2011; 96–103 therapy and/or bendamustine in patients with
65.•• Woyach JA, Johnson AJ, Byrd JC. The B-cell receptor previously treated B-cell malignancies. J Clin Oncol.
signaling pathway as a therapeutic target in CLL. 2011;29:3064. abstr.
Blood. 2012;120:1175–84. 75. O’Brien SM, Cunningham CC, Golrnkov AK, et al.
This is the most up-to date state of the art review addressing Phase I to II multicenter study of oblimersen sodium,
the subject of therapeutic intervention utilizing the B-cell a Bcl-2 antisense oligonucleotide, in patients with
receptor pathway. advanced chronic lymphocytic leukemia. J Clin
66.•• Wiestner A. Emerging role of kinase-targeted Oncol. 2005;23:7697–702.
strategies in chronic lymphocytic leukemia. Blood. 76. O’Brien S, Moore JO, Boyd TE, et al. Randomized
2012;120:4684–91. phase III trial of fludarabine plus cyclophosphamide
This is the most complete recent review of this group of with or without oblimersen sodium (Bcl-2 antisense)
agents which are likely to play a significant role in the in patients with relapsed or refractory chronic lym-
treatment of CLL. phocytic leukemia. J Clin Oncol. 2007;25:1114–20.
67. Al-Ameri AM, Badoux X, Ferrajoli A, et al. Phase II study 77. Roberts AW, Seymour JF, Brown JR, et al. An
of dasatinib in patients with relapsed chronic lym- ongoing phase 1/2a study of ABT-263;
phocytic leukemia. Blood. 2010;116:4488. abstract. pharmacokinetics (PK), safety and anti-tumor
68. Amrein PC, Attar EC, Takvorian T, et al. Phase II activity in patients(pts) with relapsed or refractory
study of dasatinib in relapsed or refractory chronic chronic lymphocytic leukemia (CLL). Blood.
lymphocytic leukemia. Clin Cancer Res. 2009;114:883. abstract.
2011;17:2977–86. 78. Kipps TJ, Swinnen LJ, Wierda WG, et al. Navitoclax
69.•• de Rooij MFM, Kuil A, Geest CR, et al. The clinically (ABT-263) plus Fludarabine/Cyclophosphamide/
active BTK inhibitor PCI-32765 targets B-cell Rituximab (FCR) or Bendamustine/Rituximab (BR):
Leukemia (JP Dutcher, Section Editor)

A Phase 1 Study In Patients with Relapsed/Refractory during remission induction in CLL and early results of such
Chronic Lymphocytic Leukemia (CLL). Blood. studies are beginning to appear.
2011;118:3904. abstract. 85. Andritsos LA, Johnson AJ, Lozanski G, et al.
79. Stolz C, Hess G, Hahnel PS, et al. Targeting Bcl-2 Higher doses of lenalidomide are associated with
family proteins modulates the sensitivity of B-cell unacceptable toxicity including life-threatening
lymphoma to rituximab-induced apoptosis. Blood. tumor flare in patients with chronic lymphocytic
2008;112:3312–21. leukemia. J Clin Oncol. 2008;26:2519–25.
80. Chen R, Guo L, Chen Y. et al.:Homoharringtonine 86. Ferrajoli A, Badoux XC, O’Brien S, et al. The
reduced Mcl-1 expression and induced apoptosis in combination of lenalidomide and rituximab induces
chronic lymphocytic leukemia. Blood. complete and partial responses in patients with
2011;117:156–64. relapsed and refractory chronic lymphocytic
81. Kumar SK, Rajkumar SV, Dispienzieri A, et al. Im- leukemia. Blood. 2010;116:1395. abstract.
proved survival in multiple myeloma and the impact 87. Ferrajoli A, Falchi L, O’Brien S, et al. Combination of
of novel therapies. Blood. 2008;111:2516–20. ofatumumab and lenalidomide in patients with re-
82. List A, Dewald G, Bennett J, et al. Lenalidomide in lapsed chronic lymphocytic leukemia (CLL): Results
the myelodysplastic syndrome with chromosome 5q of a phase II trial. Blood. 2012;117:720. abstract.
deletion. N Engl J Med. 2006;355:1456–65. 88. Brown JR, Abramson J, Hochberg E, et al. A phase I
83. Dredge K, Horsfall R, Robinson S, et al. Orally study of lenilidomide in combination with
administered lenalidomide is anti-angiogenic in vivo fludarabine and rituximab in previously untreated
and inhibits endothelial cell migration and Akt CLL/SLL. Leukemia. 2010;24:1972–5.
phosphorylation in vitro. Microvasc Res. 89. Edelmann J, Holzmann K, Miller F, et al. High-
2005;69:56–63. resolution genomic profiling of chronic lymphocytic
84.•• Chen CI, Bergsagel L, Harminder P, et al. Single-agent leukemia reveals new recurrent genomic alterations.
lenalidomide in the treatment of previously Blood. 2012;120:4783–94.
untreated chronic lymphocytic leukemia. J Clin 90. Schuh A, Becq J, Humphray S, et al. Monitoring
Oncol. 2010;29:1175–81. chronic lymphocytic leukemia progression by whole
The use of immunomodulation agents continues to interest genome sequencing reveals heterogenous clonal
investigators, particularly as consolidation agents as well as evolution patterns. Blood. 2012;120:4191–6.

You might also like