You are on page 1of 14

The n e w e ng l a n d j o u r na l of m e dic i n e

Review Article

Dan L. Longo, M.D., Editor

Treatment of Chronic Lymphocytic


Leukemia
Jan A. Burger, M.D., Ph.D.​​

C
From the Department of Leukemia, Uni- hronic lymphocytic leukemia (CLL) cells proliferate in second-
versity of Texas M.D. Anderson Cancer ary lymphoid organs (lymph nodes and spleen), where B-cell receptor sig-
Center, Houston. Address reprint requests
to Dr. Burger at the Department of Leu- naling promotes the expansion of the monoclonal B lymphocytes. B-cell
kemia, Unit 428, University of Texas M.D. receptor signaling is the target of kinase inhibitors, which have transformed CLL
Anderson Cancer Center, Houston, TX therapy during the past decade. Bruton’s tyrosine kinase (BTK) and isoform-selec-
77030, or at ­jaburger@​­mdanderson​.­org.
tive phosphatidylinositol 3-kinase (PI3K) inhibitors disrupt B-cell receptor signal-
N Engl J Med 2020;383:460-73. ing and other circuits between CLL cells and the tissue microenvironment. BCL2
DOI: 10.1056/NEJMra1908213
Copyright © 2020 Massachusetts Medical Society.
(B-cell lymphoma 2), an antiapoptotic molecule strongly expressed in CLL cells,
has emerged as another important therapeutic target. The transition from chemo-
therapy-based regimens to new, molecularly targeted agents is based on a series
of clinical trials, most of them published in the Journal,1-10 showing a survival
benefit from BTK inhibitors (ibrutinib and acalabrutinib) and the BCL2 antagonist
venetoclax, as compared with older CLL therapies (chemotherapeutic agents,5 anti-
CD20 antibodies,2,3,11 or combinations of these agents7-10,12-14). These benefits are
profound in patients with high-risk CLL and are less pronounced in those with
low-risk CLL. However, the new treatment approaches also come with challenges
(i.e., the emergence of drug resistance, serious side effects, and high costs). Com-
bination therapies to achieve deeper remissions, allowing for fixed-duration or
intermittent treatment, will help to optimize the use of the new targeted drugs.

B ackground
CLL is the most common leukemia in adults in Western countries, with a male
predominance and an average age at diagnosis of 72 years. The disease is charac-
terized by an accumulation of monoclonal, mature, CD5+ B cells in the periph-
eral blood, bone marrow, and secondary lymphoid organs. CLL is 10 to 20 times
as common in Western countries as in Asia, suggesting that genetic factors, envi-
ronmental factors, or both influence susceptibility to the disease. Often, CLL is
diagnosed after a workup for incidental lymphocytosis. The presentation and
clinical course are highly variable, ranging from asymptomatic, indolent disease
that may never require therapy (in approximately 30% of patients) to active disease
that can lead to progressive lymphocytosis, cytopenias (anemia and thrombocyto-
penia), lymphadenopathy, hepatosplenomegaly, B symptoms (i.e., weight loss, night
sweats, and fever), fatigue, recurrent infections, or autoimmune complications.
Autoimmune complications — in most cases, autoimmune thrombocytopenia or
hemolytic anemia — can occur without other signs of CLL progression and with-
out reflecting an alteration in the natural history of the disease or the prognosis
and should be treated primarily with glucocorticoids.
The guidelines of the International Workshop on Chronic Lymphocytic Leuke-
mia (iwCLL) provide unified disease staging and guidance for the clinical manage-

460 n engl j med 383;5  nejm.org  July 30, 2020

The New England Journal of Medicine


Downloaded from nejm.org at The University of Melbourne on July 31, 2020. For personal use only. No other uses without permission.
Copyright © 2020 Massachusetts Medical Society. All rights reserved.
Treatment of Chronic Lymphocytic Leukemia

Table 1. Response Rates and Survival with Chemoimmunotherapy or Novel Agents for the Treatment of Chronic
Lymphocytic Leukemia (CLL).*

Overall Response Progression-free


Treatment Disease State (Complete Remission) Survival Overall Survival

% (%)
FCR Previously untreated CLL 20
90 (44) Median, 56.8 mo Median, 12.7 yr
BR Previously untreated CLL21 96 (31) Median, 41.7 mo 92% at 3 yr
Chlorambucil plus Previously untreated CLL 22
77.3 (22.3) Median, 29.2 mo NA
obinutuzumab
Ibrutinib Relapsed or refractory CLL19 89 (10) Median, 52 mo 55% at 7 yr
Ibrutinib Previously untreated CLL5,18 92 (30) 70% at 5 yr 83% at 5 yr
Acalabrutinib Relapsed or refractory CLL4,23 94 62% at 45 mo >90% at 16 mo
Acalabrutinib Previously untreated CLL14 86 (1) 82% at 30 mo 94% at 30 mo
Acalabrutinib plus Previously untreated CLL14 94 (13) 90% at 30 mo 95% at 30 mo
obinutuzumab
Venetoclax Relapsed or refractory CLL6 79 (20) Median, 25 mo 84% at 24 mo
Venetoclax plus Relapsed or refractory CLL 7
93.3 (26.8) 84.9% at 24 mo 92% at 24 mo
rituximab
Venetoclax plus Previously untreated CLL10 84.7 (49.5) 88.2% at 24 mo 91.8% at 24 mo
obinutuzumab

* More detailed data, including side effects, and additional data from other key CLL clinical trials are provided in Tables
S1, S2, and S3 in the Supplementary Appendix, available with the full text of this article at NEJM.org. BR denotes
bendamustine and rituximab, FCR fludarabine, cyclophosphamide, and rituximab, and NA not available.

ment of CLL.15 Patients with early-stage (low- The diagnosis of CLL is based on the presence
risk) CLL (lymphocytosis alone; Rai stage 0, Binet of lymphocytosis (>5000 lymphocytes per cubic
stage A) have an estimated median life expec- millimeter) with a typical immune phenotype on
tancy of approximately 13 years16,17 and normally flow cytometry. Bone marrow testing and radio-
are not treated. Patients with intermediate-stage logic studies generally are not required for the
CLL (lymphocytosis with lymphadenopathy, hep- initial workup or follow-up. Prognostic markers
atosplenomegaly, or both; Rai stage I or II, Binet include cytogenetic abnormalities such as del(13q),
stage B) have an estimated median life expec- del(17p), trisomy 12, and del(11q),24 as well as
tancy of 8 years and can be treated if they have the mutation status of immunoglobulin heavy-
signs of disease activity. Patients with advanced- chain variable (IGHV) genes of the B-cell recep-
stage (high-risk) CLL (lymphocytosis with lymph- tor.25,26 Patients with one or more high-risk
adenopathy, hepatosplenomegaly, or both, as well markers — del(17p), del(11q), or unmutated IGHV
as marrow infiltration–related anemia, throm- — characteristically have a shorter time to initial
bocytopenia, or both; Rai stage III or IV, Binet treatment and shorter remissions after chemo-
stage C) should always be treated; these patients therapy-based treatment than patients with the
formerly had an estimated median life expec- following markers for low-risk CLL: del(13q),
tancy of only 2 years, according to data derived trisomy 12, or mutated IGHV. To estimate the
from the period when alkylating agents consti- risk of progression and time to initial treatment
tuted the primary treatment of CLL. However, in individual patients with early-stage disease, the
survival has improved substantially with the International Prognostic Index for Chronic Lym-
more recent, widespread use of the novel agents. phocytic Leukemia (CLL-IPI) can be used, which
For example, 83% of patients with CLL remain categorizes risk (low, intermediate, high, or very
alive 5 years after starting frontline ibrutinib high) on the basis of weighted individual risk
therapy,18 and 55% of patients with relapsed or factors (status with respect to 17p deletion and
refractory CLL remain alive 7 years after starting TP53 mutation, IGHV mutation status, serum β2-
ibrutinib salvage therapy19 (Table 1). microglobulin level [<3.5 mg per liter or ≥3.5 mg

n engl j med 383;5  nejm.org  July 30, 2020 461


The New England Journal of Medicine
Downloaded from nejm.org at The University of Melbourne on July 31, 2020. For personal use only. No other uses without permission.
Copyright © 2020 Massachusetts Medical Society. All rights reserved.
The n e w e ng l a n d j o u r na l of m e dic i n e

per liter], age, and clinical stage).27 The median B- Cel l R ecep t or Signa l ing
time to initial therapy is 7 years for patients a nd the Patho gene sis of CL L
with low or intermediate risk, according to the
CLL-IPI (accounting for approximately 75% of Secondary lymphoid organs are the primary site
patients with CLL), as compared with 2 years for for CLL cell proliferation,29 which has been
patients with a high or very high risk (approxi- shown directly in patients with CLL by labeling
mately 25% of patients). proliferating CLL cells with deuterated (“heavy”)
When CLL was last reviewed in the Journal,28 water.30 In lymph nodes, CLL cells form charac-
the importance of the B-cell receptor for patho- teristic proliferation centers that mimic normal
genesis and prognostication, along with the role B-cell follicles (“pseudofollicles”) (Figs. 1 and 2).
of the tissue microenvironment, was highlighted, Here, CLL cells engage in cellular and molecular
but agents for targeting these pathways were interactions that resemble those of normal B
lacking. Since then, kinase inhibitors blocking cells expanding in germinal centers during adap-
B-cell receptor signaling and the BCL2 antago- tive immune responses.31 CLL cells interact with
nist venetoclax have been introduced and are T cells32 and monocyte-derived macrophages
fundamentally changing the CLL treatment land- (so-called nurselike cells), resulting in CLL-cell
scape. activation with a B-cell receptor and nuclear fac-

A B

C D

Figure 1. Chronic Lymphocytic Leukemia (CLL) Cells Proliferating in the Lymph-Node Microenvironment.
Panels A, B, and C show sections from CLL-affected lymph nodes at progressively higher magnifications (hematoxy-
lin and eosin). Round areas of activated and proliferating CLL cells, termed pseudofollicles (also called proliferation
centers), are surrounded by sheets of darker, resting lymphocytes. Pseudofollicles are a hallmark of CLL histopathol-
ogy. In Panel D, Ki-67 staining highlights proliferating CLL cells (brown) within the proliferation centers.

462 n engl j med 383;5 nejm.org July 30, 2020

The New England Journal of Medicine


Downloaded from nejm.org at The University of Melbourne on July 31, 2020. For personal use only. No other uses without permission.
Copyright © 2020 Massachusetts Medical Society. All rights reserved.
Treatment of Chronic Lymphocytic Leukemia

tor κΒ (ΝF-κΒ) gene signature.29 This signature ma, are not found in untreated CLL, although
is recapitulated when CLL cells are cultured in mutations in PLCG2 (the gene encoding phospho-
vitro with nurselike cells,33 suggesting that mac- lipase C gamma 2) can emerge when resistance
rophages are important for triggering B-cell re- to BTK inhibitors develops.34 Therefore, B-cell
ceptor signaling in CLL. receptor activation in CLL is normally antigen-
The precise mechanism underlying the activa- dependent, resulting from B-cell receptor en-
tion of B-cell receptor signaling in CLL remains gagement with antigens in lymph nodes and the
controversial. Activating pathway mutations, spleen. For example, B-cell receptors can bind
similar to those in diffuse large B-cell lympho- various autoantigens, including calreticulin, cyto-

A B
Resting Nurselike cell
Lymph CLL cell
node

Nurselike cell
Other
CLL cell

BTK
reticular cell BCR BCR
CXCL12
CXCR4
CXCL13 Proliferating
PROLIFERATION CENTER CLL cell BCR
CXCR5
SYK
CCL3
and CCL4 BTKi
PI3K PI3Ki
CCR1 CD40 ligand
CCR4
CLL T cell
CCR5 T cell

C Anti-CD20

CD20

BCL2 antagonist

Figure 2. B-Cell Receptor (BCR) Signaling in the Proliferation of CLL Cells.


Interactions between CLL cells and the tissue microenvironment (i.e., nonmalignant cells), such as macrophages
(also referred to as nurselike cells), T cells, and reticular stromal cells result in activation and proliferation of the
CLL cells (Panel A). BCR signaling in CLL cells is activated in secondary lymphatic organs by soluble or cell-surface
antigens, which can be autoantigens (e.g., calreticulin on nurselike cells) (Panel B). Homotypic interactions between
BCR molecules (right upper corner) result in autonomous BCR signaling. BCR activation causes downstream signal-
ing through spleen tyrosine kinase (SYK), Bruton’s tyrosine kinase (BTK), and phosphatidylinositol 3-kinase (PI3K),
which are the targets of the BTK inhibitors (BTKi) ibrutinib and acalabrutinib and the PI3K inhibitors (PI3Ki) idelalis-
ib and duvelisib. SYK, BTK, and PI3K also transmit signaling from chemokine receptors, such as CXCR4 and CXCR5,
which regulate tissue homing and retention of CLL cells. In response to BCR signaling, CLL cells secrete the che-
moattractants CCL3 and CCL4, which attract T cells for cognate interactions, as seen in CLL proliferation centers.
This loop is also effectively inhibited by the kinase inhibitors (BTKi and PI3Ki). T cells, in turn, promote CLL-cell pro-
liferation through CD40–CD40 ligand interactions. CLL cells constitutively express high levels of the antiapoptotic
protein BCL2, which is localized in the outer membrane of mitochondria (Panel C). BCL2 is targeted by the BCL2
antagonist venetoclax, a BH2 mimetic, resulting in CLL-cell apoptosis. The surface antigen CD20 is the target of
antibodies (rituximab, ofatumumab, and obinutuzumab), which are commonly administered in combination with
chemotherapy or targeted therapies. CCR denotes C-C chemokine receptor, and CXCL C-X-C motif chemokine ligand.

n engl j med 383;5 nejm.org July 30, 2020 463


The New England Journal of Medicine
Downloaded from nejm.org at The University of Melbourne on July 31, 2020. For personal use only. No other uses without permission.
Copyright © 2020 Massachusetts Medical Society. All rights reserved.
The n e w e ng l a n d j o u r na l of m e dic i n e

skeletal nonmuscle myosin heavy chain IIA, vi- tor–activated CLL cells and function as plasma
mentin, IgG, DNA, or apoptotic cells. Microbial biomarkers in patients with CLL treated with
antigens, such as lipopolysaccharides, or β-(1,6)- kinase inhibitors that target BTK38 or PI3K.39
glucan, a fungal antigen, can also activate CLL
B-cell receptors and, in turn, promote CLL pro- IGH V Mu tat ion S tat us a nd
liferation.35 In addition, B-cell receptor activation S ter eo t y ped B- Cel l R ecep t or s
in CLL can occur when two CLL B-cell receptors
engage each other,36 resulting in autonomous The mutation status of IGHV genes influences
signaling (Fig. 2B). Signaling downstream of the the natural history of CLL. IGHV mutation analy-
B-cell receptor activates kinases (LYN, spleen tyro- sis distinguishes patients whose CLL cells ex-
sine kinase [SYK], BTK, and PI3K) (Fig. 3). Among press mutated IGHV with 2% or greater deviation
these kinases, BTK has emerged as the most from the IGHV germline sequence (who gener-
successful therapeutic target. Downstream sig- ally have indolent disease) from patients with
naling through extracellular signal-regulated ki- unmutated CLL with less than 2% deviation25,26
nase (ERK) and NF-κB results in transcriptional (who present with more active and treatment-
activation. An example is activation of the T-cell resistant disease). These differences in clinical
chemoattractants CCL3 and CCL4, which are behavior have been linked to differences in B-cell
secreted at high concentrations by B-cell recep- receptor responsiveness. Unmutated CLL clones

BCR
Antigen
Chemokine
Integrins receptors
(e.g., CXCR4) CD19
β α CD79
CD20 BTKi PI3Ki
A B CCL3
CCL4
CELL MEMBRANE
LYN SYK BTK PI3K

PLCG2 Mutations in
BTKi-resistant CLL
Ca2+
PKC
ERK
NF-κB

Transcription

CYTOPLASM NUCLEUS

Figure 3. BCR Signaling, Chemokine Receptors, and Adhesion Molecules.


Antigen binding to the BCR results in the formation of clusters of surface molecules, which include the BCR, CD19,
CD20, CXCR4, and LYN. BCR signals are transmitted through the BCR complex–associated molecules CD79A and
CD79B, resulting in activation of immunoreceptor tyrosine-based activation motif (ITAM) residues on the cytoplas-
mic tails of CD79A and CD79B. This, in turn, activates SYK, BTK, PI3K, and phospholipase C gamma 2 (PLCG2). BTK
and PLCG2 mutations are often found in BTKi-resistant CLL. Further responses downstream include calcium mobili-
zation, activation of protein kinase C (PKC), and the extracellular signal-regulated kinase (ERK)–mitogen-activated
protein kinase (MAPK) and nuclear factor κB (NF-κB) pathways. This signaling response promotes activation of
transcription in the nucleus and favors CLL-cell survival and proliferation. Through IgM signaling–induced down-
regulation of the transcriptional repressor BCL6,37 the T-cell chemoattractants CCL3 and CCL4 are induced and be-
come secreted by CLL cells. SYK, BTK, and PI3K, the targets of the kinase inhibitors, also transmit signals from
chemokine receptors and adhesion molecules (integrins), which regulate CLL-cell migration and homing to the sec-
ondary lymphatic organs. Conversely, kinase inhibitors targeting these molecules disrupt the function of chemokine
receptors and integrins, resulting in defective homing and the mobilization of CLL cells from secondary lymphatic
organs to the peripheral blood, causing redistribution lymphocytosis.

464 n engl j med 383;5 nejm.org July 30, 2020

The New England Journal of Medicine


Downloaded from nejm.org at The University of Melbourne on July 31, 2020. For personal use only. No other uses without permission.
Copyright © 2020 Massachusetts Medical Society. All rights reserved.
Treatment of Chronic Lymphocytic Leukemia

respond to various autoantigens, triggering ro- treated with lower-intensity regimens (Fig. 4).
bust B-cell receptor signaling, whereas mutated The addition of anti-CD20 antibodies to chemo-
CLL cells display more selective binding to re- therapy resulted in prolonged survival with both
stricted antigens and less active B-cell receptor low-intensity regimens22,44 and higher-intensity
signaling. Consequently, mutated CLL clones re- regimens,20 and chemoimmunotherapy therefore
main stable or expand at a slower rate than clones became standard therapy before the advent of
from patients with unmutated CLL. Patients with the new agents (Table 1). The combination of
unmutated IGHV also used to have a worse prog- fludarabine, cyclophosphamide, and rituximab
nosis — due to shorter remissions and shorter (FCR)20,45 was commonly used for younger, fit
survival after chemotherapy-based treatments patients; bendamustine combined with rituximab
— than patients with mutated IGHV.25,26 The (BR)21,46 was commonly used for elderly patients;
negative prognostic effect of unmutated IGHV is and chlorambucil with anti-CD20 antibodies was
overcome when patients are treated with the used for elderly patients with coexisting condi-
new targeted agents. Long-term follow-up of pa- tions.22,44 However, a series of randomized tri-
tients receiving ibrutinib as frontline therapy als2,5,7-10,12 showed better survival with the new
showed that the survival of patients with unmu- agents (kinase inhibitors and venetoclax), leading
tated CLL was excellent and similar to the sur- to a decline in the use of chemoimmunotherapy.
vival of those with mutated IGHV.18 FCR was the first regimen to induce complete
The discovery of virtually identical (stereo- remissions in a large proportion of patients45
typed) B-cell receptor sequences in unrelated and to improve overall survival (median progres-
patients with CLL solidified the concept that sion-free survival, 56.8 months)20,47 (Table 1).
common antigens drive the development and Long-term analyses revealed that low-risk pa-
progression of CLL. Approximately 30% of pa- tients often have durable remissions after treat-
tients can be grouped into CLL subgroups on the ment with FCR,48 whereas higher-risk patients
basis of shared immunoglobulin sequence mo- typically have shorter remissions.47 The shortest
tifs,40 with distinct clinical behavior and func- remissions are associated with del(17p) (5-year
tional differences in terms of B-cell receptor progression-free survival, 15.3%), followed by
activation.41 del(11q) (5-year progression-free survival, 31.4%)
Aggressive CLL is not universally associated and unmutated CLL (5-year progression-free
with unmutated IGHV, and IGHV sequencing may survival, 33.1%).47 Achievement of undetectable
not always be available. Testing CLL cells for minimal residual disease (MRD) (defined as <1 CLL
expression of CD38 and the zeta chain–associ- cell per 10,000 cells in the blood or marrow)
ated protein kinase 70 (ZAP-70) with the use of after treatment with FCR is also a strong predic-
flow cytometry provides valuable additional in- tor of progression-free survival, which may prompt
formation. Positivity for CD3825 or ZAP-7042,43 can earlier treatment discontinuation and can attenu-
function as a surrogate marker for IGHV muta- ate the negative prognostic effect of unmutated
tion status (i.e., positivity for CD38 or ZAP-70 IGHV genes.48 FCR treatment is associated with a
often — but not always — is associated with plateau in the progression-free survival curve for
unmutated IGHV and indicates an unfavorable patients with CLL and mutated IGHV genes
prognosis). (progression-free survival at 12.8 years, 53.9%).48
Promising modifications of the FCR regimen
incorporate new agents (ibrutinib49 and obinutuz­
T r e atmen t A pproache s
umab, an anti-CD20 antibody used instead of
Chemoimmunotherapy rituximab50), reduce the number of fludarabine
Before the introduction of the new agents, CLL and cyclophosphamide cycles, or both.50,51
therapy was based mostly on chemotherapy BR, another widely used regimen, is less effec-
comprising alkylating agents (chlorambucil, cyclo- tive but also less myelosuppressive than FCR.21
phosphamide, and bendamustine), nucleoside ana- When BR was compared with a new agent (ibru-
logues (fludarabine, pentostatin, and cladribine), tinib8 or venetoclax7), BR-treated patients had
and glucocorticoids. Younger, fit patients typi- significantly shorter progression-free survival.
cally received more intensive therapy, whereas Similarly, randomized trials comparing the new
elderly patients with coexisting conditions were agents with chlorambucil plus anti-CD20 anti-

n engl j med 383;5  nejm.org  July 30, 2020 465


The New England Journal of Medicine
Downloaded from nejm.org at The University of Melbourne on July 31, 2020. For personal use only. No other uses without permission.
Copyright © 2020 Massachusetts Medical Society. All rights reserved.
The n e w e ng l a n d j o u r na l of m e dic i n e

Glucocorticoids Fludarabine Chemoimmunotherapy


and alkylating plus cyclophos- with FCR induces
agents introduced phamide high CR rates
bendamustine

Nucleoside Progression-free survival


analogs benefit from anti-CD20
(fludarabine) mAb obinutuzumab plus
introduced chlorambucil vs. rituximab
plus chlorambucil or
FCR regimen
Rai and Binet chlorambucil alone
improves overall
staging
survival

1950 1960 1970 1980 1990 2000 2005 2010 2011 2012 2013 2014 2015 2016 2017 2018 2019
SYK inhibitor Progression-free
fostamatinib survival benefit
active in CLL from venetoclax
plus rituximab
vs. BR in RR CLL

Better progression-
BTK inhibitor ibrutinib active free survival with
in refractory, high-risk CLL ibrutinib with or
without rituximab
vs. BR in untreated
PI3Kδ inhibitor idelalisib active
CLL
in refractory, high-risk CLL

Progression-free survival and overall survival benefit from Progression-free survival and
ibrutinib vs. chlorambucil in untreated CLL overall survival benefit from
ibrutinib plus rituximab
vs. FCR in untreated CLL
BCL2 antagonist venetoclax and
second generation BTKi acalabrutinib
Venetoclax plus ibrutinib
active in refractory, high-risk CLL
induces high rates
of CR in untreated CLL

Progression-free benefit from


venetoclax plus obinutuzumab
vs. chlorambucil plus
obinutuzumab in untreated CLL

Figure 4. Milestones in Clinical CLL Research.


Shown in yellow above the time line are traditional chemotherapy-based approaches to treatment. Systemic therapy with alkylating
agents and glucocorticoids for patients with CLL was introduced in the 1950s, and alkylators, especially chlorambucil, given alone or
with glucocorticoids, remained widely used, even though they did not improve survival or result in deep remissions. Nucleoside ana-
logues became effective combination partners for alkylators in the 1990s, and with the addition of rituximab in the early 2000s, the con-
cept of chemoimmunotherapy was established. Fludarabine, cyclophosphamide, and rituximab (FCR) became the first therapeutic regi-
men to provide a survival benefit. Shown in green below the time line are targeted therapies. Since 2010, kinase inhibitors targeting BTK
and PI3K isoforms, the BCL2 antagonist venetoclax, and newer CD20 antibodies, such as obinutuzumab, have been approved. In ran-
domized trials, BTK inhibitors and venetoclax improve survival, as compared with chemotherapy-based regimens. CR denotes complete
remission, BR bendamustine plus rituximab, mAb monoclonal antibody, and RR refractory or relapsed.

bodies showed significantly longer progression- Early clinical testing revealed a characteristic
free survival with the BTK inhibitors12,14 or with pattern of response to the kinase inhibitors.1,2,52
venetoclax plus obinutuzumab.10 First, as compared with patients who have other
B-cell cancers, patients with CLL have the high-
Kinase Inhibitors est response rates and the most durable remis-
For the treatment of CLL, kinase inhibitors that sions. Second, kinase inhibitors typically mobilize
target the B-cell receptor signaling kinases SYK, CLL cells to move from the tissues to the periph-
BTK, and PI3K isoforms are normally adminis- eral blood, causing rapid shrinkage of enlarged
tered daily as oral agents until toxicity or CLL lymph nodes, along with a transient increase in
progression precludes further administration. peripheral-blood CLL cell counts (“redistribution

466 n engl j med 383;5  nejm.org  July 30, 2020

The New England Journal of Medicine


Downloaded from nejm.org at The University of Melbourne on July 31, 2020. For personal use only. No other uses without permission.
Copyright © 2020 Massachusetts Medical Society. All rights reserved.
Treatment of Chronic Lymphocytic Leukemia

lymphocytosis”). Third, in contrast to chemoim- had shorter remissions (7-year progression-free


munotherapy, kinase inhibitors generally are not survival, 34%).19 Common toxic effects were di-
myelosuppressive and work well in patients with arrhea, myalgia, arthralgia, and bruising,1 which
high-risk and chemotherapy-refractory CLL. normally did not require treatment interruptions
or adjustments. Minor bleeding and bruising are
BTK Inhibitors common with BTK inhibitors and are due to
Ibrutinib is the first-in-class BTK inhibitor, which inhibition of BTK-mediated platelet adhesion.
forms a covalent bond to cysteine 481 in BTK. A Infectious complications occur most frequently
nonreceptor tyrosine kinase, BTK plays a central at the beginning of ibrutinib therapy and decline
role in B-cell receptor signaling and B-cell devel- over time.61
opment. BTK was named after Dr. Ogden Bruton, The RESONATE trial showed a major survival
a pediatrician, who first described a hereditary benefit with ibrutinib as compared with the anti-
immunodeficiency syndrome, X-linked agam- CD20 antibody ofatumumab, leading to Food and
maglobulinemia, in the 1950s.53 Patients with Drug Administration (FDA) approval of ibrutinib
X-linked agammaglobulinemia lack blood B cells in 2014 for relapsed or refractory CLL.3 The ef-
and immunoglobulins as a result of functional ficacy and safety of ibrutinib as frontline therapy
null BTK mutations.54 was established in the RESONATE-2 trial, which
After triggering B-cell receptors, BTK is acti- showed significantly prolonged progression-free
vated by upstream kinases (LYN and SYK). BTK survival (70% at 5 years)18 and overall survival
also transmits signals from chemokine receptors with ibrutinib as compared with chlorambucil.5
and adhesion molecules (integrins),55,56 which Response rates and improvements in hemoglo-
organize B-cell migration and homing to the bin levels and platelet counts were greater with
lymphatic tissues (Fig. 4). Functional inhibition ibrutinib. Arterial hypertension and atrial fibril-
of these homing receptors is the basis for redis- lation were documented as additional side ef-
tribution lymphocytosis,38,57 which causes the fects.5 The Alliance A041202 trial showed longer
white-cell count to increase by a factor of 2 to progression-free survival with ibrutinib, alone or
3 or more during the first weeks of therapy in in combination with rituximab, than with BR in
most patients with CLL. This lymphocytosis, previously untreated patients with CLL.8 The ad-
which is asymptomatic, typically resolves during dition of rituximab to ibrutinib, as compared
the first months of therapy (although it can per- with ibrutinib alone, did not prolong progres-
sist for more than 12 months) and is not a sign sion-free survival in this trial8 or in another ran-
of resistance or a suboptimal response.58 On the domized trial.62 The iLLUMINATE trial, which
basis of the estimated tissue leukemia-cell burden compared ibrutinib plus obinutuzumab with
and correlations with redistribution lymphocy- chlorambucil plus obinutuzumab,12 also showed
tosis, approximately a quarter of tissue CLL cells markedly longer progression-free survival in the
are mobilized to the peripheral blood when ibrutinib group. Finally, the National Clinical
ibrutinib treatment is started.59 Conversely, a Trials Network E1912 trial showed improved
majority of tissue CLL cells undergo cell death progression-free and overall survival with ibruti-
without redistribution to the blood and normal­ nib plus rituximab as compared with FCR.9
ly without signs of tumor lysis. Serial measure- Acalabrutinib, a second-generation, covalent
ments of leukemia-cell proliferation and death BTK inhibitor, has higher BTK selectivity than
rates after deuterium labeling in patients with ibrutinib; for example, acalabrutinib does not
CLL showed abrogation of cell proliferation and inhibit epidermal growth factor receptor (EGFR),
a robust increase in cell death once ibrutinib interleukin-2–inducible T-cell kinase (ITK), or
therapy was started.60 TEC, which are tyrosine kinases that are at least
The first trial of ibrutinib in patients with partially inhibited by ibrutinib. A phase 1–2 study
relapsed or refractory CLL and a small cohort of showed high response rates among patients with
previously untreated patients1 showed high re- relapsed or refractory CLL.4 The side effects of
sponse rates, mostly partial responses,1 and very acalabrutinib, including headache, diarrhea, up-
durable responses in patients without previous per respiratory tract infection, fatigue, nausea,
treatment (7-year progression-free survival, 83%), arthralgia, hypertension, and atrial fibrillation,
whereas patients with relapsed or refractory CLL are similar to those of ibrutinib (except for aca-

n engl j med 383;5  nejm.org  July 30, 2020 467


The New England Journal of Medicine
Downloaded from nejm.org at The University of Melbourne on July 31, 2020. For personal use only. No other uses without permission.
Copyright © 2020 Massachusetts Medical Society. All rights reserved.
The n e w e ng l a n d j o u r na l of m e dic i n e

labrutinib-related headaches), but their frequen- treated with ibrutinib.65 Venetoclax can be con-
cy may be lower as a result of higher selectivity sidered as an alternative for patients at risk for
for BTK. cardiac arrhythmias.
Acalabrutinib received FDA approval for the Fungal infections, especially aspergillus in-
treatment of CLL (frontline treatment and fection, are another safety concern with ibruti-
treatment of relapsed CLL) on the basis of two nib. The incidence of aspergillus infection as-
randomized trials, ELEVATE-TN and ASCEND. sociated with ibrutinib therapy is lower among
ELEVATE-TN, which evaluated acalabrutinib, with patients with CLL than among patients with
or without obinutuzumab, as compared with central nervous system lymphoma. Predominant
obinutuzumab plus chlorambucil, showed sig- sites of infection are the lungs and central ner-
nificantly longer progression-free survival in the vous system, and an early onset after the start of
acalabrutinib groups.14 The ASCEND trial, which ibrutinib therapy is characteristic.66 General anti-
compared acalabrutinib with idelalisib (a PI3K fungal prophylaxis is not warranted in patients
inhibitor) plus rituximab or BR,13 also showed with CLL, but particular attention to the risk of
superior progression-free survival in the acala- fungal infection, especially during the first
brutinib group. Adverse events of interest with months of therapy, is recommended for high-
acalabrutinib in the ASCEND trial (median fol- risk patients (i.e., patients receiving concomitant
low-up, 16.1 months) were atrial fibrillation (in glucocorticoid therapy, those who have received
5% of patients), bleeding (in 26%), grade 3 or multiple lines of prior therapy, and those with
higher infections (in 15%), and secondary can- diabetes or liver disease).
cers (in 14%). In the ELEVATE-TN trial (median Patients with CLL already have an increased
follow-up, 28 months), adverse events associated risk of bleeding complications, as compared
with acalabrutinib included atrial fibrillation (in with healthy age-matched people, and treatment
3% of patients in the acalabrutinib-alone group with ibrutinib and other BTK inhibitors increas-
and 4% in the acalabrutinib–obinutuzumab es the frequency of low-grade bleeding, especially
group), grade 3 or higher bleeding (in 2% of bruising. The bleeding is due to inhibition of
patients in both groups), and grade 3 or higher BTK-dependent platelet aggregation. An integrat-
hypertension (in 3% of patients in the acalabru- ed analysis of 15 ibrutinib trials revealed that
tinib-alone group and 2% in the acalabrutinib– the rate of low-grade bleeding was higher with
obinutuzumab group). An ongoing randomized ibrutinib than with the comparator (35% vs. 15%).
trial will ultimately determine whether acalabru- Major hemorrhage was also more common with
tinib differs significantly from ibrutinib with ibrutinib than with the comparator (occurring in
respect to the side effect profile or efficacy. 4.4% of patients vs. 2.8%), but after adjustment
Zanubrutinib, another covalent BTK inhibi- for the longer duration of therapy with ibrutinib,
tor, is also more BTK-selective than ibrutinib is. the incidence of major hemorrhage was similar
On the basis of encouraging phase 1–2 data,63 (3.2 and 3.1 events per 1000 person-months,
CLL registration trials are ongoing. respectively).67 In view of these effects, BTK in-
hibitors should be withheld perioperatively, and
Cardiac Arrhythmias, Fungal Infections, patients receiving anticoagulant therapy should
and Bleeding with BTK Inhibitors be monitored for signs of bleeding.
Several randomized studies showed an increased
incidence of atrial fibrillation in ibrutinib-treat- PI3K Inhibitors
ed patients.5,64 In a pooled analysis of ibrutinib- Idelalisib, an oral reversible inhibitor of PI3Kδ,
treated patients, the incidence of atrial fibrilla- inhibits the signaling of the B-cell receptor and
tion was 6.5% after 16.6 months, as compared CLL homing receptors.68 Like the BTK inhibi-
with 1.6% for comparator therapy, and increased tors, idelalisib causes redistribution lymphocyto-
to 10.4% after 36 months.64 Most patients with sis,39 which is shortened by the addition of anti-
atrial fibrillation received medical management, CD20 antibodies, such as rituximab. In a
including anticoagulant therapy, and did not need randomized phase 3 trial, idelalisib plus ritux-
to permanently discontinue ibrutinib. There have imab significantly prolonged the rate of progres-
also been rare reports of ventricular arrhythmias sion-free survival (93% at 24 weeks; median
and sudden death from cardiac causes in patients survival, 20.3 months69) and overall survival, as

468 n engl j med 383;5  nejm.org  July 30, 2020

The New England Journal of Medicine


Downloaded from nejm.org at The University of Melbourne on July 31, 2020. For personal use only. No other uses without permission.
Copyright © 2020 Massachusetts Medical Society. All rights reserved.
Treatment of Chronic Lymphocytic Leukemia

compared with placebo plus rituximab.2 Because approval as frontline treatment and treatment
of autoimmune side effects (hepatitis, colitis, and of relapsed CLL. The durability of responses in
pneumonitis), infectious side effects (pneumo- venetoclax-treated patients is associated with the
cystis pneumonia and cytomegalovirus infection), depth of remission; patients with a complete
and lower efficacy than the BTK inhibitors,13 response and remissions with undetectable MRD
idelalisib is not the first-choice kinase inhibitor have the longest remissions, whereas patients
for CLL therapy, but it is a valuable alternative with incomplete remissions, bulky disease, prior
for patients in whom BTK inhibitors are associ- therapy with a BTK inhibitor, or more than three
ated with unacceptable side effects. prior lines of therapy have shorter remissions.73
Duvelisib is a dual PI3K inhibitor that targets Venetoclax is an attractive alternative if BTK
PI3Kδ and PI3Kγ. In the DUO trial, duvelisib was inhibitors have unacceptable side effects or if
compared with ofatumumab.11 Longer progres- limited-duration therapy is preferred. In addition,
sion-free survival (13.3 months vs. 9.9 months) venetoclax is active in CLL that has developed
and higher response rates supported the FDA ap- resistance to kinase inhibitors (ibrutinib or idelal-
proval of duvelisib for relapsed or refractory CLL. isib), with an overall response in 65% of patients
and a median progression-free survival of 24.7
BCL2 Antagonist months.74 Given the unrestricted approval of
Venetoclax is an orally administered, potent se- ibrutinib, acalabrutinib, and venetoclax for CLL
lective inhibitor of BCL2.70,71 BCL2 is located in therapy in the United States, sequencing of these
the outer mitochondrial membrane, where it agents is becoming an important topic. Cross-
inhibits proapoptotic proteins and supports cell trial comparisons are problematic, given the dif-
survival. BCL2 overexpression in CLL has been ferent regimens (long-term continuous adminis-
linked to the deletion of miR-15a and miR-16-1, tration vs. time-limited administration), but so
two microRNAs located at chromosome 13q14.3.72 far, no major differences in survival have been
During the early development of venetoclax, noted between ibrutinib and venetoclax as front-
cases of severe tumor lysis syndrome occurred. line treatment for CLL.
Therefore, slower dose escalation, with weekly In a phase 2 study, ibrutinib was administered
dose ramp-up and strict monitoring for the tu- in combination with venetoclax for 24 months.75
mor lysis syndrome, was implemented, with After 12 months, an impressive 88% of patients
proven safety in subsequent patients.6,7,10 had a complete response, and 61% had undetect-
The phase 1 trial of venetoclax in patients with able MRD. However, unlike patients treated with
relapsed or refractory CLL showed high response chemoimmunotherapy76 or venetoclax,73 for whom
rates across all subgroups of patients with CLL the depth of remission closely correlates with
and a median progression-free survival of 25 the duration of the treatment response, patients
months.6 Besides the tumor lysis syndrome, neu- treated with a BTK inhibitor have an excellent
tropenia (but not thrombocytopenia) was a com- outcome without necessarily having a deep re-
mon side effect. In the MURANO trial, fixed- mission,58 provided they can receive long-term
duration therapy with venetoclax, given for 24 treatment that is associated with acceptable side
cycles (28 days per cycle) and combined with effects. The appeal of combination therapy in
rituximab during the first 6 cycles, significantly which BTK inhibitors are administered with
improved survival, as compared with BR (2-year venetoclax, anti-CD20 antibodies, or both is the
progression-free survival rate, 84.9% vs. 36.3%).7 potential for treatment of limited duration.
The CLL14 trial of the German CLL Study Group Long-term follow-up of the combined-treatment
also showed a major survival benefit with fixed- trials and randomized trials will determine
duration venetoclax, given for 12 cycles and whether deeper remissions achieved by combin-
combined with obinutuzumab during cycles 1 ing BTK inhibitors with other agents translate
through 6, as compared with chlorambucil plus into any survival benefit, as compared with BTK
obinutuzumab in elderly, previously untreated inhibitor monotherapy administered indefinitely.
patients with CLL (2-year progression-free sur- The anticipated randomized CLL17 trial of the
vival rate, 88.2% vs. 64.1%).10 On the basis of the German CLL Study Group, comparing ibrutinib
CLL14 and MURANO data, venetoclax in combi- as the standard of care with venetoclax plus
nation with anti-CD20 antibodies received FDA obinutuzumab or venetoclax plus ibrutinib, is

n engl j med 383;5  nejm.org  July 30, 2020 469


The New England Journal of Medicine
Downloaded from nejm.org at The University of Melbourne on July 31, 2020. For personal use only. No other uses without permission.
Copyright © 2020 Massachusetts Medical Society. All rights reserved.
The n e w e ng l a n d j o u r na l of m e dic i n e

expected to address this important question. ed rather than up-regulated AID,83 which is more
Given the shorter follow-up with venetoclax and consistent with the antiproliferative activity of
lack of prospective data about salvage therapy the kinase inhibitors.
for patients with venetoclax-resistant CLL, BTK Venetoclax resistance can be manifested as
inhibitors normally are used before venetoclax recurrent CLL or, less frequently, as transforma-
in most patients treated outside of clinical trials. tion to diffuse large B-cell lymphoma (Richter’s
transformation) and has been linked to BCL2
Resistance to the Novel Agents mutations (G101V) that reduce the binding of
Primary resistance to BTK inhibitors is highly venetoclax to BCL2, often with additional BCL2
unusual in CLL. Redistribution lymphocytosis, mutations,84 emergence of clones with a complex
an expected on-target effect during the first karyotype, mutations in BTG1 (the B-cell trans-
months of therapy, should not be confused with location gene) and aberrations of CDKN2A or
disease progression. In patients who truly have CDKN2B (the cyclin-dependent kinase genes,
disease progression soon after the initiation of which are more common with Richter’s trans-
kinase inhibitor therapy, Richter’s transforma- formation), and overexpression of the prosur-
tion (histologic progression to diffuse large B-cell vival proteins BCL-XL and MCL1.85 Prospective
lymphoma), which normally occurs in less than data on the management of venetoclax-resistant
5% of patients with CLL, or alternative diagnoses CLL are needed. Although resistance is a common
need to be considered. Secondary resistance (i.e., reason for the failure of venetoclax treatment in
treatment failure after an initial response to a patients with relapsed CLL,73 ibrutinib therapy,
BTK inhibitor) can be due to Richter’s transfor- especially when used in early lines of treatment,
mation, which characteristically occurs early dur- is more likely to fail because of adverse toxic
ing the first year of therapy,77,78 presumably from effects.77,86
progression of preexisting high-grade clones. CLL
progression during treatment with ibrutinib char- Cellular Therapy
acteristically occurs later in high-risk patients.1,79 Although the use of BTK inhibitors and veneto-
Resistance to BTK inhibitors is often associ- clax has significantly improved survival, patients
ated with BTK mutations at the ibrutinib binding with high-risk CLL continue to be at risk for
site (C481S), activating mutations in PLCG2, or progressive disease due to the development of
both.34,78,80 BTK mutations reduce the binding of resistance. Therefore, allogeneic hematopoietic
kinase inhibitors and therefore their efficacy, stem-cell transplantation should be considered
whereas PLCG2 mutations result in BTK-indepen- in selected younger patients with high-risk dis-
dent pathway activation.34 Moreover, del(8p) and ease — those with del(17p), TP53 mutations, or
additional driver mutations have been described both and a complex karyotype — especially if
in ibrutinib-resistant CLL.81 In a few selected they have previously received chemoimmunother-
cases tested with droplet-microfluidic technology apy and subsequently had a relapse.6,10,61 The tim-
and growth kinetic analyses, ibrutinib-resistant ing of transplantation has substantially changed
subclones were already detectable at low levels in with the introduction of the new agents. Given
pretreatment samples,81 suggesting that BTK the high response rates with the new agents and
inhibitor therapy can cause clonal selection and the significant transplantation-related morbidity
expansion. It has been proposed that kinase in- and mortality,87 patients generally receive a BTK
hibitor therapy causes genomic instability through inhibitor, venetoclax, or both before they are
up-regulation of activation-induced cytidine de- considered to be candidates for hematopoietic
aminase (AID), which could pave the way for stem-cell transplantation. In high-risk patients,
drug resistance or other cancers, on the basis of remissions with ibrutinib can be durable when it
data from mouse models and human B-cell is given as frontline therapy, but remissions are
lines.82 Fortunately, AID up-regulation by idelal- generally shorter after prior exposure to chemo-
isib or ibrutinib could not be reproduced in therapy. For example, the estimated 5-year pro-
primary CLL cells (i.e., serial CLL cell samples gression-free survival rate among patients with
from ibrutinib-treated patients) or in CLL cells CLL and del(17p) or TP53 mutations was 74%
induced in vitro to express AID by CD40 ligand when ibrutinib was used as frontline therapy but
plus interleukin-4 stimulation. Under these more only 19% when it was used for the treatment of
physiologic conditions, ibrutinib down-regulat- relapsed CLL.88

470 n engl j med 383;5  nejm.org  July 30, 2020

The New England Journal of Medicine


Downloaded from nejm.org at The University of Melbourne on July 31, 2020. For personal use only. No other uses without permission.
Copyright © 2020 Massachusetts Medical Society. All rights reserved.
Treatment of Chronic Lymphocytic Leukemia

Clinical progress with autologous CD19 chi- acalabrutinib) induce durable remissions in the
meric antigen receptor (CAR) T-cell therapy in majority of patients, which are, however, mostly
CLL has been decelerated because of low re- partial. Hence, long-term treatment is required
sponse rates and short remissions.89 Recently, a when a BTK inhibitor is used as a single agent,
trial of CD19-targeting CAR T cells (at a 1:1 ratio
a plan that can be complicated by toxic effects
of CD4 and CD8 cells), together with ibrutinib, and the emergence of resistance. The BCL2 an-
in patients with ibrutinib-refractory CLL showed tagonist venetoclax, a more recent addition to
more promising results.90 This approach was more the therapeutic armamentarium, is typically given
active, inducing remissions with undetectable for a limited time, together with anti-CD20 anti-
MRD in the marrow in 61%, with a 12-month bodies, to induce deep remissions, but resistant
progression-free survival rate of 59% and an clones can emerge and clinical relapses occur,
overall survival rate of 86% among patients with especially in high-risk patients. The use of che-
undetectable MRD; it also had a better side-­ moimmunotherapy is steadily declining, given
effects profile, with less severe cytokine releasethe better side-effect profiles and improved sur-
syndrome. HLA-mismatched, CD19-directed CAR– vival with the novel agents. An exception is the
natural killer cells derived from cord blood may small group of younger, fit patients with low-
become another option for cellular therapy, given risk CLL (i.e., mutated IGHV), who often stay in
promising early safety and efficacy results.91 Be-remission for more than 10 years after treatment
cause approximately half of the patients with CLL with the FCR regimen. For such patients, FCR
who undergo allogeneic hematopoietic stem-cell treatment remains an alternative to the newer
transplantation have long-term remissions and agents. Clearly, the new agents have already im-
survival,87 allogeneic transplantation remains theproved the prognosis and quality of life for many
standard cellular therapy approach at this time. patients with CLL, especially those with high-
risk CLL. The challenge for this new decade is
to capitalize on the early success of the novel
C onclusions
agents with regimens that reduce the duration of
Basic and translational research underscored the drug exposure and the associated risks of toxic
importance of the B-cell receptor,28 the CLL mi- effects and resistance, as well as treatment costs.
croenvironment,92 and antiapoptotic proteins70 in Dr. Burger reports receiving grant support and advisory board
fees from Pharmacyclics, grant support, advisory board fees, and
the biology of CLL, and we now can look back at lecture fees from Gilead, advisory board fees from AstraZeneca,
a decade of transformative clinical research in and lecture fees and travel support from Janssen. No other poten-
CLL, with approval of drugs that target these key tial conflict of interest relevant to this article was reported.
Disclosure forms provided by the author are available with the
disease pathways. Within a short time, the BTK full text of this article at NEJM.org.
and PI3K inhibitors and venetoclax overturned I thank M.D. Anderson’s Moon Shot Program in CLL, the CLL
CLL treatment and replaced chemotherapy-based Global Research Foundation, Dr. Andreas Rosenwald for pro-
viding photomicrographs of lymph nodes in patients with CLL,
treatments for most patients who have this leu- and my family and colleagues for all their encouragement and
kemia. The BTK inhibitors (i.e., ibrutinib and support.

References
1. Byrd JC, Furman RR, Coutre SE, et al. with chronic lymphocytic leukemia. N Engl 10. Fischer K, Al-Sawaf O, Bahlo J, et al.
Targeting BTK with ibrutinib in relapsed J Med 2015;​373:​2425-37. Venetoclax and obinutuzumab in patients
chronic lymphocytic leukemia. N Engl J 6. Roberts AW, Davids MS, Pagel JM, with CLL and coexisting conditions. N Engl
Med 2013;​369:​32-42. et al. Targeting BCL2 with venetoclax in J Med 2019;​380:​2225-36.
2. Furman RR, Sharman JP, Coutre SE, relapsed chronic lymphocytic leukemia. 11. Flinn IW, Hillmen P, Montillo M,
et al. Idelalisib and rituximab in relapsed N Engl J Med 2016;​374:​311-22. et al. The phase 3 DUO trial: duvelisib vs
chronic lymphocytic leukemia. N Engl J 7. Seymour JF, Kipps TJ, Eichhorst B, ofatumumab in relapsed and refractory
Med 2014;​370:​997-1007. et al. Venetoclax–rituximab in relapsed or CLL/SLL. Blood 2018;​132:​2446-55.
3. Byrd JC, Brown JR, O’Brien S, et al. refractory chronic lymphocytic leukemia. 12. Moreno C, Greil R, Demirkan F, et al.
Ibrutinib versus ofatumumab in previ- N Engl J Med 2018;​378:​1107-20. Ibrutinib plus obinutuzumab versus chlo-
ously treated chronic lymphoid leukemia. 8. Woyach JA, Ruppert AS, Heerema NA, rambucil plus obinutuzumab in first-line
N Engl J Med 2014;​371:​213-23. et al. Ibrutinib regimens versus chemoim- treatment of chronic lymphocytic leukae-
4. Byrd JC, Harrington B, O’Brien S, munotherapy in older patients with untreat­ mia (iLLUMINATE): a multicentre, ran-
et al. Acalabrutinib (ACP-196) in relapsed ed CLL. N Engl J Med 2018;​379:​2517-28. domised, open-label, phase 3 trial. Lancet
chronic lymphocytic leukemia. N Engl J 9. Shanafelt TD, Wang XV, Kay NE, et al. Oncol 2019;​20:​43-56.
Med 2016;​374:​323-32. Ibrutinib–rituximab or chemoimmuno- 13. Ghia P, Pluta A, Wach M, et al. ASCEND:
5. Burger JA, Tedeschi A, Barr PM, et al. therapy for chronic lymphocytic leukemia. Phase III, randomized trial of acalabruti-
Ibrutinib as initial therapy for patients N Engl J Med 2019;​381:​432-43. nib versus idelalisib plus rituximab or

n engl j med 383;5  nejm.org  July 30, 2020 471


The New England Journal of Medicine
Downloaded from nejm.org at The University of Melbourne on July 31, 2020. For personal use only. No other uses without permission.
Copyright © 2020 Massachusetts Medical Society. All rights reserved.
The n e w e ng l a n d j o u r na l of m e dic i n e

bendamustine plus rituximab in relapsed 26. Hamblin TJ, Davis Z, Gardiner A, stereotypy in chronic lymphocytic leuke-
or refractory chronic lymphocytic leuke- Oscier DG, Stevenson FK. Unmutated Ig mia. Leukemia 2017;​31:​282-91.
mia. J Clin Oncol 2020 May 27 (Epub V(H) genes are associated with a more ag- 41. Maity PC, Bilal M, Koning MT, et al.
ahead of print). gressive form of chronic lymphocytic leu- IGLV3-21*01 is an inherited risk factor for
14. Sharman JP, Egyed M, Jurczak W, et al. kemia. Blood 1999;​94:​1848-54. CLL through the acquisition of a single-
Acalabrutinib with or without obinutuzu­ 27. International CLL-IPI Working Group. point mutation enabling autonomous BCR
mab versus chlorambucil and obinutuzmab An international prognostic index for pa- signaling. Proc Natl Acad Sci U S A 2020;​
for treatment-naive chronic lymphocytic tients with chronic lymphocytic leukae- 117:​4320-7.
leukaemia (ELEVATE TN): a randomised, mia (CLL-IPI): a meta-analysis of individ- 42. Crespo M, Bosch F, Villamor N, et al.
controlled, phase 3 trial. Lancet 2020;​395:​ ual patient data. Lancet Oncol 2016;​17:​ ZAP-70 expression as a surrogate for im-
1278-91. 779-90. munoglobulin-variable-region mutations
15. Hallek M, Cheson BD, Catovsky D, 28. Chiorazzi N, Rai KR, Ferrarini M. in chronic lymphocytic leukemia. N Engl
et al. iwCLL Guidelines for diagnosis, in- Chronic lymphocytic leukemia. N Engl J J Med 2003;​348:​1764-75.
dications for treatment, response assess- Med 2005;​352:​804-15. 43. Rassenti LZ, Huynh L, Toy TL, et al.
ment, and supportive management of CLL. 29. Herishanu Y, Pérez-Galán P, Liu D, ZAP-70 compared with immunoglobulin
Blood 2018;​131:​2745-60. et al. The lymph node microenviron- heavy-chain gene mutation status as a
16. Rai KR, Sawitsky A, Cronkite EP, Cha- ment promotes B-cell receptor signal- predictor of disease progression in chronic
nana AD, Levy RN, Pasternack BS. Clini- ing, NF-kappaB activation, and tumor lymphocytic leukemia. N Engl J Med 2004;​
cal staging of chronic lymphocytic leuke- proliferation in chronic lymphocytic leu- 351:​893-901.
mia. Blood 1975;​46:​219-34. kemia. Blood 2011;​117:​563-74. 44. Hillmen P, Robak T, Janssens A, et al.
17. Binet JL, Auquier A, Dighiero G, et al. 30. Herndon TM, Chen S-S, Saba NS, et al. Chlorambucil plus ofatumumab versus
A new prognostic classification of chron- Direct in vivo evidence for increased pro- chlorambucil alone in previously untreat-
ic lymphocytic leukemia derived from a liferation of CLL cells in lymph nodes ed patients with chronic lymphocytic leu-
multivariate survival analysis. Cancer 1981;​ compared to bone marrow and peripheral kaemia (COMPLEMENT 1): a randomised,
48:​198-206. blood. Leukemia 2017;​31:​1340-7. multicentre, open-label phase 3 trial. Lan-
18. Burger JA, Barr PM, Robak T, et al. 31. Burger JA, Wiestner A. Targeting B cet 2015;​385:​1873-83.
Long-term efficacy and safety of first-line cell receptor signalling in cancer: preclin- 45. Keating MJ, O’Brien S, Albitar M, et al.
ibrutinib treatment for patients with CLL/ ical and clinical advances. Nat Rev Cancer Early results of a chemoimmunotherapy
SLL: 5 years of follow-up from the phase 3 2018;​18:​148-67. regimen of fludarabine, cyclophospha-
RESONATE-2 study. Leukemia 2020;​ 34:​ 32. Patten PEM, Buggins AG, Richards J, mide, and rituximab as initial therapy for
787-98. et al. CD38 expression in chronic lympho- chronic lymphocytic leukemia. J Clin On-
19. Byrd JC, Furman RR, Coutre SE, et al. cytic leukemia is regulated by the tumor col 2005;​23:​4079-88.
Ibrutinib treatment for first-line and re- microenvironment. Blood 2008;​111:​5173- 46. Fischer K, Cramer P, Busch R, et al.
lapsed/refractory chronic lymphocytic leu- 81. Bendamustine combined with rituximab
kemia: final analysis of the pivotal phase 33. Burger JA, Quiroga MP, Hartmann E, in patients with relapsed and/or refrac-
1b/2 PCYC-1102 study. Clin Cancer Res et al. High-level expression of the T-cell tory chronic lymphocytic leukemia: a
2020 March 24 (Epub ahead of print). chemokines CCL3 and CCL4 by chronic multicenter phase II trial of the German
20. Hallek M, Fischer K, Fingerle-Rowson lymphocytic leukemia B cells in nurselike Chronic Lymphocytic Leukemia Study
G, et al. Addition of rituximab to fludara- cell cocultures and after BCR stimulation. Group. J Clin Oncol 2011;​29:​3559-66.
bine and cyclophosphamide in patients Blood 2009;​113:​3050-8. 47. Fischer K, Bahlo J, Fink AM, et al.
with chronic lymphocytic leukaemia: a ran- 34. Woyach JA, Furman RR, Liu T-M, et al. Long-term remissions after FCR chemo­
domised, open-label, phase 3 trial. Lancet Resistance mechanisms for the Bruton’s immunotherapy in previously untreated
2010;​376:​1164-74. tyrosine kinase inhibitor ibrutinib. N Engl patients with CLL: updated results of the
21. Eichhorst B, Fink A-M, Bahlo J, et al. J Med 2014;​370:​2286-94. CLL8 trial. Blood 2016;​127:​208-15.
First-line chemoimmunotherapy with ben­ 35. Hoogeboom R, van Kessel KPM, 48. Thompson PA, Tam CS, O’Brien SM,
damustine and rituximab versus fludara- Hochstenbach F, et al. A mutated B cell et al. Fludarabine, cyclophosphamide, and
bine, cyclophosphamide, and rituximab chronic lymphocytic leukemia subset that rituximab treatment achieves long-term
in patients with advanced chronic lym- recognizes and responds to fungi. J Exp disease-free survival in IGHV-mutated
phocytic leukaemia (CLL10): an interna- Med 2013;​210:​59-70. chronic lymphocytic leukemia. Blood 2016;​
tional, open-label, randomised, phase 3, 36. Dühren-von Minden M, Übelhart R, 127:​303-9.
non-inferiority trial. Lancet Oncol 2016;​ Schneider D, et al. Chronic lymphocytic 49. Davids MS, Brander DM, Kim HT, et al.
17:​928-42. leukaemia is driven by antigen-indepen- Ibrutinib plus fludarabine, cyclophospha-
22. Goede V, Fischer K, Busch R, et al. dent cell-autonomous signalling. Nature mide, and rituximab as initial treatment
Obinutuzumab plus chlorambucil in pa- 2012;​489:​309-12. for younger patients with chronic lympho-
tients with CLL and coexisting condi- 37. Ten Hacken E, Sivina M, Kim E, et al. cytic leukaemia: a single-arm, multicen-
tions. N Engl J Med 2014;​370:​1101-10. Functional differences between IgM and tre, phase 2 trial. Lancet Haematol 2019;​
23. Byrd JC, Wierda WG, Schuh A, et al. IgD signaling in chronic lymphocytic leu- 6(8):​e419-e428.
Acalabrutinib monotherapy in patients kemia. J Immunol 2016;​197:​2522-31. 50. Jain N, Thompson PA, Burger JA, et al.
with relapsed/refractory chronic lympho- 38. Ponader S, Chen S-S, Buggy JJ, et al. Ibrutinib, fludarabine, cyclophosphamide,
cytic leukemia: updated phase 2 results. The Bruton tyrosine kinase inhibitor PCI- and obinutuzumab (iFCG) for first-line
Blood 2019;​135:​1204-13. 32765 thwarts chronic lymphocytic leuke- treatment of IGHV-mutated CLL and with-
24. Döhner H, Stilgenbauer S, Benner A, mia cell survival and tissue homing in vitro out Del(17p)/mutated TP53. Blood 2019;​
et al. Genomic aberrations and survival in and in vivo. Blood 2012;​119:​1182-9. 134:​Suppl:​357. abstract.
chronic lymphocytic leukemia. N Engl J 39. Brown JR, Byrd JC, Coutre SE, et al. 51. Dartigeas C, Van Den Neste E, Léger J,
Med 2000;​343:​1910-6. Idelalisib, an inhibitor of phosphati- et al. Rituximab maintenance versus ob-
25. Damle RN, Wasil T, Fais F, et al. Ig V dylinositol 3-kinase p110δ, for relapsed/ servation following abbreviated induc-
gene mutation status and CD38 expres- refractory chronic lymphocytic leukemia. tion with chemoimmunotherapy in elder-
sion as novel prognostic indicators in Blood 2014;​123:​3390-7. ly patients with previously untreated
chronic lymphocytic leukemia. Blood 1999;​ 40. Stamatopoulos K, Agathangelidis A, chronic lymphocytic leukaemia (CLL 2007
94:​1840-7. Rosenquist R, Ghia P. Antigen receptor SA): an open-label, randomised phase 3

472 n engl j med 383;5  nejm.org  July 30, 2020

The New England Journal of Medicine


Downloaded from nejm.org at The University of Melbourne on July 31, 2020. For personal use only. No other uses without permission.
Copyright © 2020 Massachusetts Medical Society. All rights reserved.
Treatment of Chronic Lymphocytic Leukemia

study. Lancet Haematol 2018;​ 5(2):​e82- in patients taking ibrutinib. Blood 2017;​ in chronic lymphocytic leukemia. J Clin
e94. 129:​2581-4. Oncol 2017;​35:​1437-43.
52. Friedberg JW, Sharman J, Sweeten- 66. Ruchlemer R, Ben-Ami R, Bar-Meir M, 79. Thompson PA, O’Brien SM, Wierda
ham J, et al. Inhibition of Syk with fosta- et al. Ibrutinib-associated invasive fungal WG, et al. Complex karyotype is a stron-
matinib disodium has significant clinical diseases in patients with chronic lympho- ger predictor than del(17p) for an inferior
activity in non-Hodgkin lymphoma and cytic leukaemia and non-Hodgkin lym- outcome in relapsed or refractory chronic
chronic lymphocytic leukemia. Blood 2010;​ phoma: an observational study. Mycoses lymphocytic leukemia patients treated with
115:​2578-85. 2019;​62:​1140-7. ibrutinib-based regimens. Cancer 2015;​
53. Ponader S, Burger JA. Bruton’s tyro- 67. Brown JR, Moslehi J, Ewer MS, et al. 121:​3612-21.
sine kinase: from X-linked agammaglob- Incidence of and risk factors for major 80. Ahn IE, Underbayev C, Albitar A, et al.
ulinemia toward targeted therapy for B-cell haemorrhage in patients treated with Clonal evolution leading to ibrutinib re-
malignancies. J Clin Oncol 2014;​ 32:​ ibrutinib: an integrated analysis. Br J Hae- sistance in chronic lymphocytic leukemia.
1830-9. matol 2019;​184:​558-69. Blood 2017;​129:​1469-79.
54. Smith CIE. From identification of the 68. Hoellenriegel J, Meadows SA, Sivina 81. Burger JA, Landau DA, Taylor-Weiner
BTK kinase to effective management of M, et al. The phosphoinositide 3′-kinase A, et al. Clonal evolution in patients with
leukemia. Oncogene 2017;​36:​2045-53. delta inhibitor, CAL-101, inhibits B-cell chronic lymphocytic leukaemia develop-
55. Spaargaren M, Beuling EA, Rurup receptor signaling and chemokine net- ing resistance to BTK inhibition. Nat
ML, et al. The B cell antigen receptor con- works in chronic lymphocytic leukemia. Commun 2016;​7:​11589.
trols integrin activity through Btk and Blood 2011;​118:​3603-12. 82. Compagno M, Wang Q, Pighi C, et al.
PLCgamma2. J Exp Med 2003;​198:​1539- 69. Sharman JP, Coutre SE, Furman RR, Phosphatidylinositol 3-kinase δ blockade
50. et al. Final results of a randomized, phase increases genomic instability in B cells.
56. de Gorter DJJ, Beuling EA, Kersse- III study of rituximab with or without Nature 2017;​542:​489-93.
boom R, et al. Bruton’s tyrosine kinase idelalisib followed by open-label idelalisib 83. Morande PE, Sivina M, Uriepero A,
and phospholipase Cgamma2 mediate in patients with relapsed chronic lympho- et al. Ibrutinib therapy downregulates AID
chemokine-controlled B cell migration cytic leukemia. J Clin Oncol 2019;​37:​1391- enzyme and proliferative fractions in chron-
and homing. Immunity 2007;​26:​93-104. 402. ic lymphocytic leukemia. Blood 2019;​133:​
57. de Rooij MFM, Kuil A, Geest CR, et al. 70. Hanada M, Delia D, Aiello A, Stadt- 2056-68.
The clinically active BTK inhibitor PCI- mauer E, Reed JC. bcl-2 Gene hypometh- 84. Blombery P, Thompson ER, Nguyen T,
32765 targets B-cell receptor- and chemo- ylation and high-level expression in B-cell et al. Multiple BCL2 mutations cooccur-
kine-controlled adhesion and migration chronic lymphocytic leukemia. Blood 1993;​ ring with Gly101Val emerge in chronic
in chronic lymphocytic leukemia. Blood 82:​1820-8. lymphocytic leukemia progression on
2012;​119:​2590-4. 71. Korz C, Pscherer A, Benner A, et al. venetoclax. Blood 2020;​135:​773-7.
58. Woyach JA, Smucker K, Smith LL, et al. Evidence for distinct pathomechanisms 85. Thijssen R, Roberts AW. Venetoclax in
Prolonged lymphocytosis during ibruti- in B-cell chronic lymphocytic leukemia lymphoid malignancies: new insights, more
nib therapy is associated with distinct and mantle cell lymphoma by quantitative to learn. Cancer Cell 2019;​36:​341-3.
molecular characteristics and does not expression analysis of cell cycle and apop- 86. Mato AR, Nabhan C, Thompson MC,
indicate a suboptimal response to thera- tosis-associated genes. Blood 2002;​99:​4554- et al. Toxicities and outcomes of 616 ibru-
py. Blood 2014;​123:​1810-7. 61. tinib-treated patients in the United States:
59. Wodarz D, Garg N, Komarova NL, et al. 72. Cimmino A, Calin GA, Fabbri M, et al. a real-world analysis. Haematologica 2018;​
Kinetics of CLL cells in tissues and blood miR-15 and miR-16 Induce apoptosis by 103:​874-9.
during therapy with the BTK inhibitor targeting BCL2. Proc Natl Acad Sci U S A 87. Krämer I, Stilgenbauer S, Dietrich S,
ibrutinib. Blood 2014;​123:​4132-5. 2005;​102:​13944-9. et al. Allogeneic hematopoietic cell trans-
60. Burger JA, Li KW, Keating MJ, et al. 73. Roberts AW, Ma S, Kipps TJ, et al. Ef- plantation for high-risk CLL: 10-year fol-
Leukemia cell proliferation and death in ficacy of venetoclax in relapsed chronic low-up of the GCLLSG CLL3X trial. Blood
chronic lymphocytic leukemia patients on lymphocytic leukemia is influenced by dis- 2017;​130:​1477-80.
therapy with the BTK inhibitor ibrutinib. ease and response variables. Blood 2019;​ 88. Ahn IE, Farooqui MZH, Tian X, et al.
JCI Insight 2017;​2(2):​e89904. 134:​111-22. Depth and durability of response to ibru-
61. O’Brien S, Furman RR, Coutre S, et al. 74. Jones JA, Mato AR, Wierda WG, et al. tinib in CLL: 5-year follow-up of a phase 2
Single-agent ibrutinib in treatment-naïve Venetoclax for chronic lymphocytic leu- study. Blood 2018;​131:​2357-66.
and relapsed/refractory chronic lympho- kaemia progressing after ibrutinib: an 89. Fraietta JA, Lacey SF, Orlando EJ, et al.
cytic leukemia: a 5-year experience. Blood interim analysis of a multicentre, open- Determinants of response and resistance
2018;​131:​1910-9. label, phase 2 trial. Lancet Oncol 2018;​19:​ to CD19 chimeric antigen receptor (CAR)
62. Burger JA, Sivina M, Jain N, et al. Ran- 65-75. T cell therapy of chronic lymphocytic leu-
domized trial of ibrutinib vs ibrutinib 75. Jain N, Keating M, Thompson P, et al. kemia. Nat Med 2018;​24:​563-71.
plus rituximab in patients with chronic Ibrutinib and venetoclax for first-line treat- 90. Gauthier J, Hirayama AV, Purushe J,
lymphocytic leukemia. Blood 2019;​133:​ ment of CLL. N Engl J Med 2019;​380:​ et al. Feasibility and efficacy of CD19-
1011-9. 2095-103. targeted CAR T cells with concurrent
63. Tam CS, Trotman J, Opat S, et al. 76. Strati P, Keating MJ, O’Brien SM, et al. ibrutinib for CLL after ibrutinib failure.
Phase 1 study of the selective BTK inhibi- Eradication of bone marrow minimal re- Blood 2020;​135:​1650-60.
tor zanubrutinib in B-cell malignancies sidual disease may prompt early treat- 91. Liu E, Marin D, Banerjee P, et al. Use
and safety and efficacy evaluation in CLL. ment discontinuation in CLL. Blood 2014;​ of CAR-transduced natural killer cells in
Blood 2019;​134:​851-9. 123:​3727-32. CD19-positive lymphoid tumors. N Engl J
64. Brown JR, Moslehi J, O’Brien S, et al. 77. Maddocks KJ, Ruppert AS, Lozanski Med 2020;​382:​545-53.
Characterization of atrial fibrillation ad- G, et al. Etiology of ibrutinib therapy dis- 92. Burger JA, Ghia P, Rosenwald A, Ca-
verse events reported in ibrutinib random- continuation and outcomes in patients ligaris-Cappio F. The microenvironment
ized controlled registration trials. Haema- with chronic lymphocytic leukemia. JAMA in mature B-cell malignancies: a target
tologica 2017;​102:​1796-805. Oncol 2015;​1:​80-7. for new treatment strategies. Blood 2009;​
65. Lampson BL, Yu L, Glynn RJ, et al. 78. Woyach JA, Ruppert AS, Guinn D, et al. 114:​3367-75.
Ventricular arrhythmias and sudden death BTKC481S-mediated resistance to ibrutinib Copyright © 2020 Massachusetts Medical Society.

n engl j med 383;5  nejm.org  July 30, 2020 473


The New England Journal of Medicine
Downloaded from nejm.org at The University of Melbourne on July 31, 2020. For personal use only. No other uses without permission.
Copyright © 2020 Massachusetts Medical Society. All rights reserved.

You might also like