You are on page 1of 18

REVIEWs

Chronic lymphocytic leukaemia:


from genetics to treatment
Francesc Bosch   1,2,3* and Riccardo Dalla-​Favera3
Abstract | Chronic lymphocytic leukaemia (CLL), the most frequent type of leukaemia in adults,
is a lymphoproliferative disorder that is characterized by the expansion of monoclonal, mature
CD5+CD23+ B cells in the peripheral blood, secondary lymphoid tissues and bone marrow. CLL is
an incurable disease with a heterogeneous clinical course, for which the treatment decision still
relies on conventional parameters (such as clinical stage and lymphocyte doubling time).
During the past 5 years, relevant advances have been made in understanding CLL biology.
Indeed, substantial progress has been made in the identification of the putative cell of origin of
CLL , and comprehensive studies have dissected the genomic, epigenomic and transcriptomic
landscape of CLL. Advances in clinical management include improvements in our understanding
of the prognostic value of different genetic lesions, particularly those associated with
chemoresistance and progression to highly aggressive forms of CLL , and the advent of new
therapies targeting crucial biological pathways. In this Review , we discuss new insights into the
genetic lesions involved in the pathogenesis of CLL and how these genetic insights influence
clinical management and the development of new therapeutic strategies for this disease.

With an estimated incidence of 4.7 new cases per 100,000 The  modified Rai classification, which was derived
individuals, chronic lymphocytic leukaemia (CLL) is the from the original Rai staging system with five risk sub-
most common form of chronic leukaemia in the USA and groups (stages 0–IV)13, defines patients with low-​risk CLL
in Europe1. In 2017, the estimated number of newly diag- who solely display lymphocytosis, intermediate-​risk dis-
nosed cases was 20,110, representing ~1.2% of all cancers ease with palpable lymph nodes or hepatosplenomegaly,
in the USA2. By contrast, the incidence of CLL among and high-​risk disease with anaemia (haemoglobin <110 g/l)
individuals from African, Caribbean and Asian countries or thrombocytopenia (platelet count <100 × 109/l)13. The
is ~1.4 new cases per 100,000 individuals, suggesting Binet classification (Binet A–C) relies on similar physical
that genetic factors have a role in this racial disparity3,4. examination and laboratory findings12. On the basis of the
Accordingly, relatives of patients with CLL have a higher enormous advances in the biology and treatment of CLL,
risk of developing CLL and other lymphoproliferative dis- these historical staging systems have been complemented
orders than the general population, although the basis for by a plethora of new prognostic and predictive parame-
this genetic susceptibility remains elusive5–7. The median ters based on CLL genetics and bio­logy for the purpose of
age at diagnosis is 70 years, with a male predominance predicting patient outcomes and response to treatment14–17.
(1.3:1) in all ethnic subgroups2,8. As life expectancy and In this Review, we focus on insights into the genetic
the use of blood testing increase, the prevalence of CLL lesions involved in the pathogenesis of CLL gained over the
1
Department of Haematology,
University Hospital Vall
and the proportion of asymptomatic patients diagnosed past 7 years, referring readers to other reviews on the role
d’Hebron, Autonomous with early stage disease will continue to increase, whereas of the tumour microenvironment18. We discuss the
University, Barcelona, Spain. the median age at diagnosis will decrease2,8. genetic lesions in CLL and their value as both prognostic
2
Laboratory of Experimental CLL is a chronic lymphoproliferative disorder9,10 indicators of clinical course and as predictive biomarkers
Haematology, Vall d’Hebron diagnosed by the presence of ≥5,000 clonal CD5+CD23+ of response to chemotherapy and targeted therapies.
Institute of Oncology (VHIO),
B lymphocytes per microlitre of peripheral blood for more
Barcelona, Spain.
than 3 months, or by the presence of its non-​leukaemic Genetics and pathogenesis of CLL
3
Institute for Cancer Genetics,
Columbia University,
variant, small lymphocytic lymphoma (SLL)9,10. CLL Emerging knowledge of CLL genetics and biology indi-
New York, NY, USA. remains an incurable disease with a heterogeneous clin- cates that CLL is not a homogeneous disease, informing
*e-​mail: fbosch@vhio.net ical course, for which prognostication is based on sim- the current understanding of the distinct cell types from
https://doi.org/10.1038/ ple and highly reproducible clinical parameters that which this malignancy can originate and the complex set of
s41571-019-0239-8 were used to develop two clinical staging systems11,12. genetic lesions that are associated with its pathogenesis.

Nature Reviews | Clinical Oncology


Reviews

Key points methylation in CLL cells is generally similar to that in


memory B cells, although CLL-​associated differences
• Next-​generation sequencing of the coding genome has identified the most common might reflect the existence of a currently unknown
somatic genetic alterations associated with the pathogenesis of chronic lymphocytic non-malignant B cell subpopulation24. Overall, the dis-
leukaemia (CLL).
tinction between IGHV-​M CLL putatively derived from
• Specific genetic alterations provide biomarkers for prognostication of the clinical GC-​experienced B cells and IGHV-​UM CLL arising
course and prediction of response to chemotherapy and targeted therapy.
from pre-​GC-naive B cells or GC-​independent mem-
• Recurrent genetic alterations identify cellular pathways that present potential ory B cells still remains a valid, although not universally
therapeutic targets. accepted, model.
• This new knowledge is informing novel treatment algorithms for the clinical Patients with IGHV-​M CLL and those with IGHV-​
management of patients with CLL. UM CLL have a highly restricted repertoire of IG genes,
suggesting a role for antigen selection in the patho-
Cell of origin genesis of the disease25,26. Approximately one-​third of
The identification of the cell of origin of any given patients with CLL, particularly those with IGHV-​UM
malignancy (that is, the non-​maligant cell from which CLL, exhibit stereotypic BCRs, which are based on
the malignant transformation originated) can be useful their complementary-​determining regions26,27. To date,
as a reference to identify the alterations in the cell that >200 different CLL stereotype subsets have been iden-
caused its malignant transformation. Over the years, tified25, which can be further subdivided into several
several cell types have been proposed as putative cells of subsets with distinct biological and clinical features28:
origin (also sometimes referred to as the ‘normal counter­ IGHV-​UM CLLs tend to express low-​affinity, poly-​
part’) of CLL on the basis of progressive increases in the reactive and self-​reactive BCRs, whereas IGHV-​M
understanding of B cell biology and differentiation. CLLs usually express oligo-​reactive and mono-​reactive
The observed presence of clonal rearrangements BCRs25. Notably, both IGHV-​M and IGHV-​UM sub-
of immunoglobulin (IG) genes along with expression of types might also rely on antigen-​independent, cell-​
specific cell surface markers has established that CLL autonomous BCR signalling, providing a potential
is derived from a mature B cell that is characterized rationale for the therapeutic targeting of the BCR
by weak expression of B cell markers (surface mem- signalling pathway29.
brane immunoglobulins (Ig), CD19 and CD20) and is Finally, the very early genetic and epigenetic events
positive for expression of CD23 (also known as FcεRII; that eventually lead to CLL have been suggested to
a B cell and dendritic cell marker) and of the CD200 occur in haematopoietic stem cells (HSCs)30. Consistent
and CD5 antigens10. The expression of CD5 led to the with this notion, in some patients with allogeneic HSC
hypothesis that CLL is derived from IgM-​s ecreting transplantation (HSCT), pre-​malignant lymphoid cells
B1 lineage of B cells that are contributors to innate were transmitted from the donor to the recipient and
immunity19,20, as opposed to the B2 lineage, which is eventually led to CLL development30. In addition, HSCs
involved in adaptive immunity. Subsequently, the dis- from patients with CLL, but not those from healthy
covery that two subgroups of patients with CLL can be donors, have been shown to engraft in immunodeficient
distinguished on the basis of the presence or absence mice and induce B cell lymphoproliferations in vivo31.
of mutations in the immunoglobulin heavy chain var- Finally, specific lesions in genes implicated in lymphoid
iable region (IGHV) genes, which encode part of the tumours (for example, NOTCH1, SF3B1 and EGR) have
B cell receptor (BCR), unequivocally indicated that been found in purified HSCs from some patients with
the IGHV-​mutated (IGHV-M) CLLs are derived from CLL32. Overall, the conclusion that the initial genetic
antigen-​experienced B cells that have transited through lesions favouring CLL development might occur in
the germinal centre (GC) of secondary lymphoid HSCs remains controversial, particularly considering
organs, the site of immunoglobulin somatic hyper­ the difficulties in purifying CLL-​free HSC populations
mutation21. Conversely, whether the IGHV-​unmutated from bone marrow32,33, and the emerging evidence that
(IGHV-​UM) CLLs are derived from pre-​GC (naive) healthy elderly individuals have clonal haematopoiesis,
B cells or GC-​independent antigen-experienced B cells which can increase the risk of haematological malignan-
remains unclear21. cies, including CLL34,35, and is associated with subsets of
Comparative gene expression profiling of CLL speci­ genetic lesions also detected in CLL.
mens and subsets of non-​transformed human B cell has Overall, the cell of origin of CLL is a subject of con-
shown that both IGHV-​M and IGHV-​UM subtypes are tinued debate, with regard to both the cell type in which
similar to CD27+ memory B cells21, suggesting that both the first genetic lesion occurs (HSCs versus mature
CLL subtypes originate from antigen-​experienced CD27+ B cells) and to the mature B cell type (pre-​GC, post-​GC
B cells, with IGHV-​M and IGHV-​UM subtypes having or GC-​independent) from which the clonal expansion
been derived from post-​GC or GC-​independent cells, of overt CLL can originate.
respectively21,22. In 2012, transcriptome analyses of sub-
sets of non-​transformed B cells suggested that IGHV-M Recurrent genetic lesions
CLLs are derived from a previously unrecognized The development of genome-​wide sequencing and
CD27+CD5+ post-​GC subset, whereas IGVH-​UM CLLs copy-​number analysis has enabled the thorough charac­
resemble CD5+CD27− IGHV-​UM-naive B cells23 (Fig. 1). terization of the genomic landscape of CLL (Fig.  2).
A 2012 study comparing the epigenetic profiles of Whole-​e xome sequencing (WES) of >1,000 CLL
patients with CLL showed that the degree of genome specimens and whole-​genome sequencing (WGS) of

www.nature.com/nrclinonc
Reviews

Early/immature Mature
• Lymphoid-lineage priming • Clonal selection • Clonal selection
• Expansion • Expansion • Transformation

MBL IGHV-M CLL

TD

GC Post-GC
B cell CD5+CD27+ • BCR signalling
B cell • T cells
• Microenvironmental factors
‘CLL HSC’ Naive
HSC B cell B cell
(?) TD
progenitor

Pre-GC
TI CD5+CD27–
B cell

MBL IGHV-UM CLL

Clonal/polyclonal Oligoclonal Monoclonal

Fig. 1 | The cellular origin of chronic lymphocytic leukaemia. The genetic and epigenetic events leading to chronic
lymphocytic leukaemia (CLL) have been suggested to occur in haematopoietic stem cells (HSCs)31, although this
hypothesis remains controversial32,33. After HSCs acquire genetic and epigenetic lesions (indicated by lightning symbols),
the nature of which are still unknown, the cellular output of these lesions could be a polyclonal expansion of B cell
progenitors. Subsequent antigenic stimulation might lead to oligoclonal selection and expansion of mature B cells.
CLL cells with mutations in the immunoglobulin heavy chain variable region genes (IGHV-​M) seem to originate from
post-germinal centre (GC) CD5+CD27+ B cells, which are most likely derived from CD5+CD27+ B cells that have undergone
the GC reaction23. Conversely , IGHV-​unmutated (IGHV-​UM) CLL seems to be derived from pre-​GC CD5+CD27− B cells,
which might arise from naive B cells or from a separate lineage of precursor B cells, probably in a T cell-​independent
process23. The evolution from these progenitors to monoclonal B lymphocytosis (MBL) and, ultimately , to overt CLL is
dictated by additional genetic and epigenetic abnormalities (indicated by lightning symbols), B cell receptor (BCR)
stimulation, microenvironmental factors and T cells. The dashed arrows indicate a hypothetical mechanism. TD,
T cell-dependent antigen; TI, T cell-​independent antigen. Figure adapted from ref.48, Springer Nature Limited.

~200 patients with CLL have revealed the presence of deleted region contains two long non-​coding RNA
~0.9 mutations per megabase (including point muta- genes (DLEU2 and DLEU1) and the microRNA gene
tions, copy number alterations and exceedingly rare cluster MIR15A–MIR16-1 (refs53–57). The pathogenetic
chromosomal translocations) and a load of ~10–30 role of these deletions has been confirmed in mice with
non-​silent events per patient, with a higher number of conditional deletions of the murine locus equivalent
somatic substitutions in IGHV-​M CLL (~2,800) than in to that of the minimal deleted region in patients with
IGHV-​UM CLL (~2,000)36–45. Overall, the mutational CLL, which developed clonal lymphoproliferations,
load in CLL is lower than in other lymphoid neoplasms recapitulating the different steps of CLL initiation and
and epithelial tumours, and is similar to that in acute progression, including monoclonal B lymphocytosis
leukaemias46,47. In this section, we summarize the major (MBL), CLL, and progression to diffuse large B cell
pathways that are recurrently altered in a sizeable pro- lymphoma (DLBCL)51,53. In vitro studies have indicated
portion of patients with CLL, and discuss their prog- that the main mechanisms by which MIR15A–MIR16-1
nostic value as individual biomarkers (Table 1). We refer exert their pathogenetic roles in B cells is through
to other reviews that provide detailed reports of the full regu­lation of the cell cycle and apoptosis, specifically
spectrum of genetic alterations in CLL48,49. by modulating the expression of genes involved in the
G0/G1–S transition (for example, inducing activation of
Regulation of the cell cycle and apoptosis. The most G1–S-​specific cyclin D2 (CCND2) and CCND351) and
frequent genetic lesions in CLL (~50–60% of patients)50 the anti-​apoptotic gene BCL2 (ref.58).
are deletions of 13q14 (del13q14) (Fig.  2; Table  1) , Since the seminal study by Döhner and colleagues50,
which are generally monoallelic (~80% of del13q14 the detection of del13q14 has been associated with the
events) and more prevalent in IGHV-​M CLL than in best prognosis among the different copy number
IGHV-​UM CLL41,50–52. These lesions are often found abnormalities (del17p, del11q and trisomy 12) (Table 1).
to be the unique cytogenetic abnormality detectable Patients with CLL and del13q14 tend to have a prolonged
in some CLLs, suggesting their role in the early stages time to first treatment (TTFT), mutations in the IGHV
of CLL development41,50,51. Despite the fact that the size of genes and prolonged overall survival (OS) compared
13q14 deletion is variable across patients, the minimal with patients with other genetic abnormalities15,50.

Nature Reviews | Clinical Oncology


Reviews

NOTCH1 BCR signalling Inflammatory Not recurrently mutated


signalling pathway
<5%
Brontictuzumab Fostamatinib BCR TLR 5–10%
Entospletinib IL-1R
RO4949097 10–20%
LY3039478 TAK659
>20%
Cerdulatinib
PF03084014
NOTCH1

γ-secretase Ibrutinib SNS-062 LYN


Acalabrutinib M7583 MYD88 IMO-8400
PLCγ2
SYK NFKBIE BIRC3 CA-4948
FBXW7 BGB-3111 DTRMWXHS
CT-1530 CC-292 BLNK TRAF2/
BTK Venetoclax
Tirabrutinib TRAF3 BA
Navitoclax X
Idelalisib PI3K NF-κB MIK665 BAK
Duvelisib KRAS (anti-MCL1)
Umbralisib NRAS AMG176
AKT (anti-MCL1) PUMA BIM
YY-20394
BRAF
RNA processing
BCL2
RPS15 mTOR MAPKs
Chromatin
modification APR-246
NXF1 Cell cycle COTI-2
XPO1 and apoptosis DNA damage response
ICN1

CDH2 CB-103 MED12


ATM p53
H2B H2A SF3B1 • Cyclin D2
• Cyclin D3 NF-κB
H4 H3 POT1

SETD2 Target
genes AZD6738
ASXL1 Selinexor BAY1895344
SL-801 del13q14 del11q del17p
Trisomy 12 (MIR15A–MIR16-1, (ATM) (TP53)
DLEU1 and DLEU2)

Fig. 2 | Cll genetic landscape and currently tested targeted drugs. The schematic depicts the complexity of the
chronic lymphocytic leukaemia (CLL) genetic landscape revealed by fluorescence in situ hybridization (FISH) and next-​
generation sequencing (NGS). The most frequent chromosomal abnormalities detected by FISH include deletions of
chromosome 13q14 (del13q14; which contains MIR15A–MIR16-1, DLEU1 and DLEU2), trisomy 12, del11q (which contains
ATM) and del17p (which contains TP53)50. Del13q14 has been implicated in the regulation of the cell cycle and apoptosis,
whereas del11q (ATM) and del17p (TP53) affect the response to DNA damage. Recurrent genetic alterations identified by
NGS include mutations in genes involved in chromatin modification (such as CHD2), nuclear factor-​κB (NF-​κB) signalling
(BIRC3 and MYD88), the NOTCH1 pathway (NOTCH1 and FBXW7), RNA processing (SF3B1 and XPO1) or the DNA damage
response (for example, ATM, TP53 and POT1)38–40. The frequencies of the genetic lesions and/or the encoded proteins are
indicated by their colour (see the key). Proteins that are not recurrently mutated in CLL are shown in blue. Targeted
therapies that are currently being tested in clinical trials involving patients with CLL are indicated as light-​grey boxes;
approved agents are in bold text. BCR , B cell receptor.

NOTCH1 signalling. The most commonly mutated gene analogous functional outcome via abnormally stabilized
detected at diagnosis in patients with CLL is NOTCH1, NOTCH1 signalling. The pathogenetic role of NOTCH1
a well-​characterized oncogene in T cell acute lympho­ in CLL might not be limited to patients harbouring gene
blastic leukaemia (T-​A LL) 59, with a prevalence of mutations, given that CLL cells in the lymph nodes display
~4–20%36,38–41 (Fig. 2; Table 1). NOTCH1 encodes a trans- frequent NOTCH1 activation independent of mutation61
membrane receptor that, upon binding to its ligand, is and that ICN1 and a NOTCH1 gene expression signature
proteolytically processed to generate the transcriptionally are detectable in ~50% of peripheral blood CLL cells lack-
active nuclear intracellular domain of NOTCH1 (ICN1), ing NOTCH1 mutations62. These findings highlight the
which transactivates genes involved in proliferation, need to revisit the role of NOTCH1 in CLL pathogenesis
metabolism and survival, including the activation of the and its prognostic and therapeutic implications.
MYC proto-​oncogene60. In contrast to the mutations in The frequency of NOTCH1 mutations in patients
T-​ALL, NOTCH1 mutations in CLL remove the PEST with CLL varies according to the clinical stage in which
domain of the NOTCH1 protein, preventing proteasomal it is analysed. Indeed, NOTCH1 lesions are very rare
degradation of ICN1 by the ubiquitin ligase F-box/WD (<5%) in patients with ultra-​stable CLL, but are present
repeat-​containing protein 7 (FBXW7)36,37,39,40,42. Of note, at diagnosis in ~12% of unselected patients with CLL63.
3% of CLLs that are devoid of NOTCH1 lesions har- The frequency of NOTCH1 mutation is increased in
bour FBXW7-inactivating mutations41, suggesting an patients with advanced-​stage CLL and in those with CLL

www.nature.com/nrclinonc
Reviews

Table 1 | Prognostic influence of Cll genetic lesions at diagnosis


genetic lesion Study number of patients Frequency TTFT 5-year additional features
(%) (months) OS (%)
Isolated del13q14 Döhner et al.50 325; 52% with early stage CLL 55a ~90b >90b • Enriched in early stages
• Enriched in IGHV-​M
NOTCH1 Rossi et al.63 637; 75% with early stage CLL 11 NA 50b • Enriched in advanced stages
mutation • Associated with trisomy 12
Weissmann et al.116 643; 0% with early stage CLL 12 42b 55b • Enriched in IGHV-​UM
Baliakas et al.64 2697; 78% with early stage CLL 7 40b NA
Nadeu et al.82 396; 84% with early stage CLL 22 <12b ~70b,c
Jeromin et al. 125
908; 0% with early stage CLL 12.3 40 b
80b
Mansouri et al.185 359; 77% with early stage CLL 5 5b ~50b
FBXW7 mutation Jeromin et al. 125
908; 0% with early stage CLL 2.5 NA NA NA
del17p or TP53 Döhner et al.50 325; 52% with early stage CLL 7a <12b 40b • Enriched in advanced stages
disruption • Enriched in IGHV-​UM
Zenz et al.71 125; 65% with early stage CLL 10d NA 40
Puente et al.40 452; 68% with early stage CLL 5 <12b NA
Rossi et al.15 637; 75% with early stage CLL 8.5 27b 50b
Baliakas et al.64 1691; 78% with early stage CLL 9 36 35b
del11q or ATM Döhner et al. 50
325; 52% with early stage CLL 18 a
13 b
68b • Enriched in advanced stages
disruption • Associated with bulky disease
Nadeu et al.82 398; 84% with early stage CLL 11 <12b NS • Associated with young age
Wierda et al.186 930; 87% with early stage CLL 9a <24b NA • Enriched in IGHV-​UM
POT1 mutation Ramsay et al.83 127; 82% with early stage CLL 3 NA NA Enriched in IGHV-​UM
SF3B1 disruption Wang et al. 37
91 ; 79% with early stage CLL
e
15 <12 b
NA • High male:female ratio
• Associated with high white
Rossi et al.63 637; 75% with early stage CLL 6.8 <12b 2.5 yearsb,f blood cell count
Baliakas et al.64 1715; 78% with early stage CLL 8 36b NA • Enriched in advanced stages
• Associated with del11q
Jeromin et al.125 1160; 0% with early stage CLL 9 40b 64b • Enriched in IGHV-​UM
Mansouri et al. 185
360; 77% with early stage CLL 3.6 3b
~55b
BIRC3 disruption Rossi et al.187 637; 75% with early stage CLL 6.2 NA 3.1 yearsb,f • Enriched in advanced stages
• Associated with del11q
Baliakas et al. 64
919; 78% with early stage CLL 2.5 32 b
NA • Enriched in IGHV-​UM
Nadeu et al.82 399; 84% with early stage CLL 4 NS NA
MYD88 mutation Martínez-​Trillos et al. 87,88
587; 81% with early stage CLL 4 NS 100b • Increased prevalence in males
• Associated with young ageg
Baliakas et al.188 1080; 78% with early stage CLL 2.2 ~79b NA • Enriched in advanced stages
Rossi et al.63 637; 75% with early stage CLL 4.1 NA 96b • Enriched in IGHV-​M
CLL , chronic lymphocytic leukaemia; NA , not available; NS, not significant; OS, overall survival; TTFT, time to first treatment. aFluorescence in situ hybridization;
b
Statistical prognostic significance of the lesion; cNot statistically significant at the subclonal level; dSanger sequencing; eTwo-​thirds untreated; fMedian survival;
g
Controversial data.

harbouring trisomy 12, and ~80% of patients harbouring the entire short arm of chromosome 17 and invariably
NOTCH1 mutations have IGHV-​UM CLL63,64 (Table 1). encompass the locus of the tumour suppressor gene
Moreover, their presence has been associated with TP53 (ref.73). Missense mutations in the second TP53
unfavourable OS (5-year OS 50–70%), even in patients allele, usually located in the DNA-​binding domain,
with IGHV-​M CLL, and with an increased risk of Richter are found in ~80% of CLLs with del17p, with an addi-
syndrome36,65,66. Of note, patients with genetic lesions tional 9% of mutations discovered using ultra-​deep
of the 3′ untranslated region of NOTCH1 tend to have next-​generation sequencing (ultra-​deep NGS)71,74–76.
a poor prognosis similar to that of patients with other Consistent with the increased genomic instability
NOTCH1 mutations40. Finally, NOTCH1-mutated CLLs caused by the inactivation of TP53, CLLs with del17p
have decreased surface expression of CD20, which might have a higher degree of genomic complexity than
partially explain why such patients derive limited benefit TP53-wild-type CLLs77.
from anti-​CD20-based therapies36,63,67–70. The prognostic implication of TP53 dysregulation
is linked to its association with resistance to DNA-​
DNA damage response. Deletions of the 17p13 chromo- damaging agents (chemotherapy or radiotherapy), and
somal region (del17p) are found at different frequencies the presence of this genetic lesion at diagnosis is associ-
in CLL depending on the clinical stage of the disease, ated with a decreased TTFT and unfavourable OS15,40,50,71
ranging from 1% in MBL to 20% in chemorefractory (Table 1) . Accordingly, TP53 inactivation is a major
disease70–72 (Fig. 2; Table 1). Typically, 17p losses involve determinant of therapeutic decisions.

Nature Reviews | Clinical Oncology


Reviews

ATM, an upstream regulator of the TP53 gene located complex36–41,86. Monoallelic BIRC3-truncating muta-
in the 11q22-23 region, is involved in deletions (del11q) tions are found at diagnosis in 2.2–4.3% of patients with
in ~10% of patients with CLL, with an increased fre- CLL and cause the removal of the C-​terminal RING fin-
quency in those with IGHV-​UM CLL with nodal ger domain of the BIRC3 protein, leading to constitutive
disease37,50,78 (Fig. 2; Table 1). In general, del11q is mon- NF-​κB activation79,84 (Fig. 2; Table 1). In addition, the
oallelic, large in size (>20 Mb) and is associated with MYD88 gene is mutated in ~3% of patients with CLL,
mutations in the remaining ATM allele in more than exclusively those with IGHV-M ​ CLL40,41 (Fig. 2; Table 1).
one-​third of patients with del11q CLL, suggesting that MYD88, a cytosolic protein adaptor involved in the
the ATM gene is the major target of these deletions78. Toll-​like receptor (TLR) pathway, has pleiotropic func-
In some patients, deletions of 11q do not involve ATM, tions in B cells, including NF-​κB activation. The most
but affect the BIRC3 gene, a negative regulator of the common recurrent MYD88 mutation (MYD88L265P) in
non-​canonical nuclear factor-​κB (NF-​κB) pathway79. CLL, which is also found in Waldenström macroglob-
Both del11q and ATM genetic lesions are found in up to ulinaemia and activated B cell (ABC)-type DLBCL,
20% of patients at diagnosis39,48,50,78, and these patients leads to increased binding of MYD88 to interleukin-1
tend to be younger, have enlarged lymph nodes at diag- receptor-​associated kinase 1 (IRAK1) and subsequent
nosis and have a shorter TTFT and OS than patients downstream activation of NF-​κ B 39. Finally, other
with del13q, trisomy 12 or those without chromosomal lesions in genes involved in the inflammatory pathway
abnormalities, particularly when ATM genetic lesions (TRAF2, TRAF3 and NFKBIE) are observed at frequen-
are biallelic78,80–82. cies <2% and are all hypothesized to lead to NF-​κB acti-
POT1 is involved in telomere protection and is vation86 (Fig. 2). However, a definitive assessment of the
mutated in 3–7% of patients with CLL83. POT1 mutations frequency of NF-​κB activation and the role of NF-​κB
alter the telomeric DNA binding domain, resulting in complexes in CLL pathogenesis is still lacking.
increased structural aberrations and chromosomal breaks Among the different genetic lesions that could acti-
that favour tumour progression83. These lesions have vate the NF-​κB signalling pathway, BIRC3 lesions have
been reported to be strongly associated with IGHV-UM been positively associated with chemorefractory dis-
CLL, SF3B1 mutations and advanced clinical stage83. ease, advanced clinical stages, IGHV-​UM disease and
an increased prevalence of del11q79. However, although
RNA processing. Recurrent mutations in different genes the presence of BIRC3 aberrations seems to be associ-
encoding spliceosome machinery have been identified ated with shorter OS, their influence on TTFT has not
in patients with CLL, with the gene encoding the splicing been proved82. The prognostic implications of MYD88
factor 3B subunit 1 (SF3B1) reported as the most fre- lesions remain controversial because of conflicting
quently mutated gene37,42,84, with a mutational frequency data, presumably owing to their low frequency. One
of ~10%37,40,41 (Fig. 2; Table 1). Although the role of SF3B1 study, mainly involving patients with early stage CLL
lesions is not yet fully understood, they might lead to and a low median age, revealed that MYD88 lesions
splicing changes affecting the structure and coding were positively associated with IGHV-​M status (90%),
potential of gene transcripts across multiple pathways, Binet A stage, and young age and had a small influence
including the DNA damage response and NOTCH1 on TTFT and, surprisingly, OS87,88. Conversely, analy­
signalling85. Along with SF3B1, genetic lesions in other sis of larger cohorts of patients with advanced-​stage
genes involved in RNA processing (for example, NFX1, CLL showed that MYD88 mutations are not preferen-
XPO1 and MED12) have also been reported, albeit at tially associated with age or prognosis64,84 and are not
lower frequencies48,85 (Fig. 2). In addition to the genetic associated with an adverse outcome in patients with
lesions affecting the spliceosome, mutations in RPS15, IGHV-​M CLL89.
a gene encoding a component of the 40S ribosomal
subunit, have been associated with aggressive CLL and Trisomy 12. Trisomy of chromosome 12 is a relevant
TP53 abnormalities1. Assuming that all of these lesions and recurrent cytogenetic abnormality found in ~15% of
in genes encoding spliceosome components function patients with CLL at diagnosis50, for which no functional
via the same mechanism, dysregulation of RNA pro- explanation has been found to date (Fig. 2). Trisomy 12
cessing would be observed in ~30% of patients, suggest- has been associated with an atypical morphology of the
ing a common pathogenetic pathway in CLL41,42,85. The lymphocytes, and its prevalence is markedly high in SLL,
frequency of genetic abnormalities in the SF3B1 gene the tumoural counterpart of CLL, in which it is found
tend to increase from ~10% in newly diagnosed patients in ~30% of patients90. Although trisomy 12 has classi-
to a higher frequency of ~17% in patients refractory to cally been considered as an intermediate-​risk genetic
fludarabine treatment64,84, and are more frequent in lesion, the co-​occurrence of NOTCH1 mutations has
patients with del11q, del13q14 or trisomy 12 (refs64,84). been associated with poor outcomes67,91. This finding is
These lesions are also more frequent in males than in line with the observation of an increased frequency of
females, and their presence correlates with a decreased trisomy 12 in patients with Richter syndrome92.
TTFT and unfavourable OS (Table 1).
Epigenetic changes in CLL
Inflammatory pathway and constitutive NF-​κB acti- Epigenetic studies, defined by the analysis of DNA-​
vation. Several lesions found in large-​scale genome modifying marks in the genome of tumour cells, can
sequencing projects in patients with CLL seem to con- provide biologically and clinically important informa-
verge on the activation of the NF-​κB transcriptional tion in two different areas. By comparing tumour and

www.nature.com/nrclinonc
Reviews

Risk factors
• TP53 aberrations
• NOTCH1 mutations
• IGHV4-39
del13q14 >50%
NOTCH1 ~40%
CDKN2A/B ~30% RS (5–10%)
del9p21 ~30%
MYC ~30%
• BCR signalling Risk factors
• Microenvironment • TP53 aberrations
• Advanced-stage • Bone marrow
CLL CLL disease RS under
involvement
Early stage Advanced stage ibrutinib (5%)
MBL • Clonally related
MYC gains
Ibrutinib
(<2 years)

TP53 >40%
del13q14 60% del13q14 60% BIRC3 ~24% CLL resistant
TP53 2% TP53 5% SF3B1 ~17% to CIT
SF3B1 4% SF3B1 8% del13q14 50% ATM ~20%
TP53 7% CIT
NOTCH1 6% NOTCH1 12%
ATM 5% ATM 10% SF3B1 21%
BIRC3 4% BIRC3 8% NOTCH1 10% BTK mutations
ATM 15% PLCG2 mutations CLL resistant
BIRC3 6% del8p to ibrutinib
ITPKB mutations (15%)
Ibrutinib MYC gains
(>2 years)

Fig. 3 | genetics of chronic lymphocytic leukaemia progression. The overall genomic mutation load seems to be
similar in monoclonal B lymphocytosis (MBL) and early stage chronic lymphocytic leukaemia (CLL), as determined in
both whole-exome sequencing and whole-​genome sequencing studies38–41. When CLL evolves from early stages to
overt disease, discrete acquisition of genetic lesions occurs, suggesting that B cell receptor (BCR) signalling and/or
the microenvironment contribute to disease progression41. After the constraints exerted by treatment with
chemoimmunotherapy (CIT) or the BCR signalling inhibitor ibrutinib, CLL cells with genetic lesions conferring drug
resistance become clonally selected (orange cells)15,41,45. Finally , CLL can transform from advanced-​stage disease to
an aggressive lymphoma termed Richter syndrome (RS), although the concurrent diagnosis of CLL and RS is rare
(reviewed in113). In most cases, the RS cells (red cells) derive from the original CLL clone (blue cells) by developing
additional genetic lesions65,66,92. NOTCH1 mutations, MYC activation and TP53, CDKN2A and CDKN2B disruptions are
characteristic genetic lesions of RS65. Patients with CLL receiving ibrutinib can progress in two different ways. First, in
approximately 5% of patients, the disease can evolve in <2 years on treatment to a RS that is clonally related to the
underlying CLL119,120. The time and clinical presentation suggest that ibrutinib selects for a pre-​existing RS clone.
Second, 15% of patients, who typically show progression after >2 years of treatment, can develop mutations associated
with resistance to ibrutinib (that is, BTKC481S, PLCG2 or ITPKB mutations)45,119,147. These mutations can be detected using
highly sensitive techniques even before the start of ibrutinib treatment (orange cells) and up to 15 months preceding
clinical progression, which is characterized by an increase in the size of the lymph nodes and a rise in lymphocyte count
and serum lactate dehydrogenase (LDH) levels119,120,147.

non-​transformed cells, epigenetic studies provide rele- Genetics of tumour progression


vant information on the putative cell of origin. In fact, Clinical and biological evidence indicates that CLL is
the comparative analysis of DNA methylation profiles preceded by a pre-​malignant stage of MBL, and can
of IGHV-​UM and IGHV-​M cells has suggested a rela- evolve in later, more aggressive stages, including trans­
tionship with naive and memory B cells, respectively24. formation into an aggressive lymphoma (Richter
Furthermore, high levels of intra-​sample methylation transformation) (Fig. 3).
heterogeneity correlate with high-​risk genetic lesions,
clonal evolution and an adverse prognosis93. Finally, MBL and the clonal evolution of CLL. In 2008, the WHO
comprehensive analyses are unveiling relationships recognized MBL as a diagnosis based on the presence of
between some epigenetic profiles and specific genetic monoclonal B cell populations in the peripheral blood
lesions (for example, MYD88 mutations or trisomy 12)94. at concentrations of up to 5,000 cells per microlitre (with
These observations suggest that CLL-​associated epi­ the phenotype of CLL, atypical CLL or non-​CLL (CD52+)
genetic changes can complement the analysis of genetic B cells) in the absence of other clinical features, such as
alterations in the characterization of disease subgroups, disease-​related symptoms, cytopenia or lymphadeno­
intraclonal heterogeneity and response to therapy, and pathies97. MBL is detectable in up to 12% of healthy adults
that they are also of direct therapeutic interest since when analysed using a high-​sensitivity eight-​colour flow
these epigenetic changes can be efficiently modulated cytometry approach to screen >5 × 106 total peripheral
by specific drugs94–96. blood B lymphocytes98, and in up to 25% of individuals

Nature Reviews | Clinical Oncology


Reviews

older than 70 years99. The updated WHO classification (in >85% of patients with Richter syndrome), but has a
distinguishes ‘high-​count’ MBL from ‘low-​count’ MBL, poorer outcome than de novo DLBCL and is character-
the latter being defined by a peripheral blood CLL count ized by chemoresistance and unfavourable OS66,92. Risk
of <500 cells per microlitre (refs10,100). High-​count MBL factors for the development of Richter syndrome include
has been recognized as a pre-​malignant state because it the presence of TP53 disruptions, NOTCH1 mutations
precedes CLL in virtually all patients101,102. In addition, and IGHV4-39 usage (reviewed in114). In patients receiv-
high-​count MBL carries mutated IGHV genes and a BCR ing BCR signalling inhibitors, the presence of tetraploid
repertoire and stereotype that resemble those of CLL103. karyotypes and TP53 aberrations have been associated
Conversely, low-​count MBL has an immunoglobulin G with an increased risk of Richter syndrome115. Activation
(IgG) genetic repertoire that is usually found in elderly of the NOTCH1 pathway is the most characteristic
individuals and that differs from those observed in genetic feature of clonally related Richter syndrome, con-
patients with IGHV-​M or IGHV-​UM CLL, suggesting sistent with the observations that NOTCH1 mutations in
that low-​count MBL is not a pre-​malignant state, but CLL at diagnosis are associated with an increased likeli-
rather a consequence of age-​related immune senes- hood of developing Richter syndrome and can be found
cence104. In addition, the distinction between high-count at subclonal levels by ultra-​deep NGS in the original CLL
and low-​count MBL is based on data from a small cohort founder clones36,63,65,116,117. Since NOTCH1 mutations are
(n = 31) and reflects a continuum of lymphocyte counts rarely acquired during CLL progression, patients with
that is associated with the risk of developing overt CLL52. this genetic lesion should be closely monitored as they
Thus, the updated International Workshop on Chronic are at risk of developing Richter syndrome and could be
Lymphocytic Leukemia (iwCLL) guidelines do not candidates for NOTCH1-targeted therapy118. In contrast
re­cognize low-​count MBL as a pathological entity and to clonally related Richter syndrome, the less common
do not recommend that affected patients are clinically clonally unrelated Richter syndromes (<15% of patients
followed105. Finally, further genomic and epidemiologi- with Richter syndrome) have a similar survival outcome
cal analysis of MBL is necessary to conclusively validate to that of de novo DLBCL and, therefore, should be con-
these notions106,107. sidered either as independent second malignancies or as
The overall genomic mutational load seems sim- divergent pathogenetic variants derived from a common
ilar in CLL and MBL, as evidenced by both WES and precursor cell66,113.
WGS studies39. However, the load of putative driver Genomic analysis has revealed that, in most patients,
genetic lesions is significantly lower in MBL than in CLL Richter syndrome is derived from the original predom-
(1.2 versus 1.7 mutations per megabase for IGHV-M inant CLL clone via a linear pathway of genetic evolu-
CLL; P = 0.0008)40,108, except when MBL is compared tion involving the development of additional genetic
with ultra-​stable CLL107. This notion was also verified in lesions65,66,92. The final genetic landscape of Richter syn-
a study specifically evaluating the prevalence of lesions drome displays similarities to as well as marked differ-
in genes such as NOTCH1, SF3B1 and TP53 in MBL, ences from de novo DLBCL. In particular, mutations that
which showed that the presence of these lesions is of prog- are commonly found in Richter syndrome and are also
nostic significance109. Intriguingly, progression from MBL found in de novo DLBCL include MYC-​activating events
to overt CLL does not seem to be accompanied by acqui- and disruption of TP53, CDKN2A and CDKN2B, which
sition of novel genetic lesions, but rather by an expansion are found collectively in ~90% of patients with Richter
of the clones that already harbour driver mutations109. syndrome65 (Fig. 3). Conversely, some lesions common
Overall, the precise genetic boundaries between MBL and in all DLBCL subgroups are rare in Richter syndrome,
CLL remain difficult to identify, but a consensus exists in including biallelic inactivation of the TNFAIP3 (also
defining del13q14, trisomy 12 and MYD88 mutations as known as A20) and PRDM1 (also known as BLIMP1)
early initiating lesions commonly present in CLL38,41,109, tumour suppressor genes, and BCL6 translocations. One
whereas TP53, ATM and SF3B1 mutations seem to be of the most distinguishing features of Richter syndrome
genetic events that occur during tumour progression41. DLBCL is a high frequency of trisomy 12, which is rare
Similar to other neoplasias, CLL is genetically hetero­ in de novo DLBCL65,66,92.
geneous and contains multiple clonal and subclonal Approximately 5–10% of patients with CLL receiv-
populations38,110,111. The interplay among these sub- ing ibrutinib develop Richter syndrome, usually within
clones along with their response to internal or external 2 years on treatment, suggesting that this drug selects for
constraints, such as the selection induced by drug treat- a pre-​existing transformed clone. Pathological data on
ment, will determine the abundance and identity of the Richter syndrome associated with ibrutinib treatment are
selected subclones and their evolution38,41,70,75,79,112 (Fig. 3). limited, and this transformation has been associated with
advanced-​stage disease with TP53 dysfunctions and is
Richter syndrome. Analogous to other low-​grade lympho­ usually clonally related to the underlying CLL and asso-
proliferations, CLL can transform into Richter syndrome, ciated with MYC copy number gains119,120. Finally, Richter
a lymphoma with an aggressive phenotype that has an syndrome has been described in some patients receiving
annual incidence of 0.5% among all patients with CLL the BCL2 inhibitor venetoclax121,122, although the exact
(reviewed in113). The WHO classification recognizes prevalence and characteristics of this transformation
two distinct pathological variants of Richter syndrome, remain to be determined. Some of these patients had
namely the DLBCL variant and the Hodgkin lym- previously characterized TP53 mutations in the CLL, and
phoma variant10. Richter syndrome-​associated DLBCL this transformation was associated with the acquisition of
is commonly clonally related to the founder CLL clone mutations in the CDKN2A, CDKN2B or SF3B1 (ref.122).

www.nature.com/nrclinonc
Reviews

Clinical implications using variables with independent prognostic value and


The extensive knowledge derived from the genomic the identification of subgroups of patients who are likely
analysis of CLL reveals marked heterogeneity in this to progress and, therefore, might need early treatment.
malignancy. Emerging evidence indicates that this hetero­ The identification of such patients is now of parti­cular
geneity can inform prognostication and predict resistance importance because the overwhelming majority of
to both chemotherapy and targeted therapy. new patients with CLL are diagnosed at early stages8.
Unfortunately, most prognostic markers have been
Genetic lesions as prognostic markers identified in patients with CLL treated with chemo­
The mutational status of the IGHV genes remains sta- immunotherapy, thus some variables (for example,
ble throughout the course of the disease and is one of IGHV mutational status) might have minor prognos-
the most important prognostic biomarkers in CLL. In tic value in patients treated with newer targeted treat-
comparison with patients with IGHV-​M CLL (~60% at ments. Thus, different prognostic indexes are needed for
diagnosis), those with IGHV-​UM CLL are characterized patients receiving these new therapies.
by the presence of high-​risk genetic lesions, an increased One integrated score enabled the identification of
propensity to undergo clonal evolution and an associa- four novel prognostic subgroups by combining fluo-
tion with a shorter TTFT and unfavourable OS38,40,123,124. rescence in situ hybridization (FISH)-based analysis
In addition, ~30% of patients carrying stereotyped BCRs of chromosomal aberrations with molecular data from
have distinct clinical outcomes, particularly in specific 1,274 patients at diagnosis15 (Table 2). Importantly, this
subsets (reviewed in ref.28), but further investigation of approach identified subgroups with a poor prognosis
the prognostic value of BCR stereotype is warranted. within previously established groups with a good prog-
Finally, surrogates of the mutational status of IGHV nosis. Thus, ~20% of the lower-​risk patients accord-
genes, such as tyrosine protein kinase ZAP-70, CD38 or ing to the FISH cytogenetic model were stratified
CD49d expression, have been found to be of prognostic into higher-risk groups by integrating the mutational
importance for TTFT and OS14. information.
The complexity and individually variable prognostic A second score, the CLL International Prognostic
value of the aforementioned genetic lesions have stimu­ Index (CLL-​IPI), was established in a large cohort
lated the development of integrated prognostic scores of patients who were included in eight clinical trials
combining multiple genetic lesions alone or in combi- evaluating chemotherapy or chemoimmunotherapy
nation with established clinical variables (Table 2). The combinations (chlorambucil, chlorambucil plus ritux-
main clinical value of these scores is the simplification imab, fludarabine, fludarabine plus cyclophosphamide
of the prognostic assessment of patients with CLL by and fludarabine plus cyclophosphamide plus rituximab),

Table 2 | Prognostic scores combining clinical and molecular variables in Cll


Study number of independent variables predicting independent variables
patients TTFT predicting OS
Rossi et al.15 1274 – • High-​risk: TP53 and/or BIRC3
mutation
• Intermediate-​risk: NOTCH1
mutation, SF3B1 mutation or
del11q
• Low-​risk: trisomy 12 or normal
cytogenetics
• Very low-​risk: del13q14
Jeromin et al.125 1160 • SF3B1 mutation • SF3B1 mutation
• IGHV-​UM • IGHV-​UM
• del11q • TP53 disruption
• Male sex
• Age
CLL-​IPI17 3472 • TP53 disruption (mutation or • TP53 disruption (mutation or
deletion) deletion)
• IGHV-​UM • IGHV-​UM
• Serum β2-microglobulin level • Serum β2-microglobulin level
>3.5 mg/l >3.5 mg/l
• Binet B or C (Rai I–IV) stage • Binet B or C (Rai I–IV) stage
• Age >65 years • Age >65 years
Nadeu et al.126 406 • IGHV-​UM • Age >65 years
• Binet B–C • del17p
• SF3B1 mutation • IGHV-​UM
• BRAF mutation • FBXW7 mutation
• ATM mutation • NOTCH1 mutation
• NOTCH1 mutation • SF3B1 mutation
• MGA mutation • TP53 mutation
CLL , chronic lymphocytic leukaemia; CLL-​IPI, Chronic Lymphocytic Leukemia International Prognostic Index; OS, overall survival;
TTFT, time to first treatment.

Nature Reviews | Clinical Oncology


Reviews

and validated in two independent datasets17 (Table 2). comprise drugs that induce DNA damage, clones and
Five parameters were predictive of OS and TTFT, namely subclones carrying genetic lesions that hamper DNA
TP53 status (considering either del17p or TP53 lesions), repair, particularly TP53 gene disruptions, are resist-
IGHV mutational status (mutated versus unmutated), ant to, and are therefore selected by, chemoimmuno­
serum β2-microglobulin levels (cut-​off >3.5 mg/l), therapy 38,75,82 (Fig.  3; Table  3) . Accordingly, several
clinical stage (Binet A versus B or C) and age (cut-​off studies have shown that clonal evolution occurs more
age >65 years). The value of this score is its ability to frequently in tumours receiving chemoimmunotherapy
predict OS, both for treatment decisions and at diag­ treatments, whereas an equilibrium exists in the clonal
nosis. Jeromin et al.125 analysed the prognostic influence architecture in untreated tumours41. This notion has
of a group of genes (NOTCH1, FBXW7, MYD88, XPO1 important therapeutic implications, although the pos-
and TP53) and their prognostic value along with age and sibility that the detection of aggressive clonal evolution
sex in 1,160 untreated patients with CLL (Table 2). They might in fact lead to early treatment cannot be excluded.
found that SF3B1 mutations, IGHV-​UM and del11q Nonetheless, sequential analysis of patients in relapse
are predictive of a short TTFT, whereas SF3B1 muta- after chemoimmunotherapy has shown that subclonal
tion, IGHV-​UM, TP53 disruption, male sex and age are evolution can follow three general patterns41,139. First,
correlated positively with a short OS duration. CLL subtypes with del13q, trisomy 12 or del11q tend to
Finally, the prognostic model developed by Nadeu remain clonally stable. Second, the frequency of TP53
et al.126 was obtained in a cohort of patients with mostly gene disruptions increases upon relapse and are associ-
early stage disease. Despite its complexity, this model has ated with inferior response rates as well as unfavourable
important features, including the observation that the progression-​free survival (PFS) and OS after treatment.
accumulation of driver alterations in addition to clonal Thus, all patients with CLL who are eligible for therapy
architecture are of prognostic significance126. Of note, should be tested for the existence of TP53 disruptions
SF3B1, but not TP53, disruptions had an independent before the initiation of chemoimmunotherapy. Third,
prognostic influence on the TTFT, but both were key CLL subtypes with SF3B1 and ATM lesions have a vari-
factors in predicting OS in the aforementioned models able evolution (increases or decreases in the abundance
by Nadeu et al.126 and Jeromin et al.125 (Table 2). of the subclones), although genetic lesions in SF3B1
Given the low frequency of genetic lesions at diag­ are associated with shorter PFS durations with minor
nosis, larger studies are warranted to reveal the real influences on response rates and OS.
influence of these genetic lesions in early stage disease. Notably, NOTCH1 abnormalities do not correlate
More importantly, until comprehensive genomic ana­ with response or OS after chemoimmunotherapy41,70,72,
lysis is widely incorporated into clinical practice, prog- except in one study70, in which the presence of NOTCH1
nostic scores should be kept as simple as possible and lesions was correlated with a lack of PFS benefit from
easy to apply. the addition of rituximab to fludarabine plus cyclophos-
phamide (thus forming the FCR regimen). This find-
Genetic lesions as predictive markers ing might be related to the observation that CLL cells
For almost 40 years, chlorambucil was the mainstay in with mutated NOTCH1 exhibit decreased expression of
the treatment of CLL, but had little influence on the sur- CD20 (ref.140) (Table 3). However, the prognostic influ-
vival outcomes of patients16. During the past 15 years, ence of the detection of the intracellular (ICN1) portion
several new treatment regimens — particularly chemo- of NOTCH1, an indicator of its constitutive activity, has
immunotherapy combinations — have been shown to not yet been assessed62.
be active in CLL, achieving deep responses and prolong- Finally, analysis of predictive markers of a poor
ing survival of patients127–132. Over the past 5 years, an response to fludarabine in patients with refractory CLL
impressive number of targeted therapies have emerged has unveiled the involvement of most of the genetic
that are changing most of the therapeutic paradigms in alterations associated with clinical progression in CLL,
CLL133–138. Some parameters (such as IGHV mutational including TP53 (27.5%), NOTCH1 (24.1%), SF3B1
status) that are useful predictive markers, defined as clin- (18.9%) and BIRC3 (15.5%)141. Remarkably, alloge-
ical or molecular features that provide information on neic HSCT is able to overcome the adverse predic-
the probability that a patient will derive benefit from a tive influence of these genetic lesions in patients with
specific treatment, for chemoimmunotherapy regimens fludarabine-​refractory CLL142, demonstrating that thera-
no longer remain valid when applied to targeted ther- pies for which the mechanism of action does not directly
apies. Hence, identification of the underlying mecha­ induce DNA damage should be preferred treatments for
nisms of therapeutic response and resistance in CLL patients with TP53 lesions (Table 3).
is of the utmost relevance. Thus, analysis of the afore-
mentioned main genetic lesions should be included in Response to targeted therapies. The BCR signalling
prospective clinical trials to enable selection of patients pathway has been targeted using several small-​molecule
who will derive the most benefit from a given treatment. inhibitors, with ibrutinib and idelalisib currently
approved for the treatment of CLL. Ibrutinib is an irre-
Response to chemoimmunotherapy. Recent genomic versible inhibitor of BTK that strongly inhibits BCR sig-
studies have unveiled the phenomenon of intratumoural nalling, leading to decreases in proliferation, adhesion
heterogeneity, characterized by the presence of partially and homing of CLL lymphocytes as well as to abroga-
genetically diverse clones and subclones that interact tion of the tumour-​protective effects from the micro­
and compete110. As chemoimmunotherapy regimens environment (reviewed in143). Owing to its mechanism

www.nature.com/nrclinonc
Reviews

Table 3 | Predictive value of genetic lesions according to type of treatment


genetic Treatment Study treatments (setting) number of ORR (%) mPFS (months) mOS (months)
lesion type patients
(% mutated)
TP53 CIT Alemtuzumab versus 297 (7.5) • Mutant: 64 versus 20a • Mutant: 10 versus 2a NR
chlorambucil (frontline)189 • WT: 83 versus 55a • WT: 15 versus 12a
Fludarabine versus fludarabine 378 (8.5) Mutant 67 versus WT Mutant 23 versus WT Mutant 30 versus WT
plus cyclophosphamide 91 (both arms) 62 (both arms)a NR (both arms)a
(frontline)72
Chlorambucil versus fludarabine 466 (7.6) Mutant 27 versus WT Mutant 5% versus WT Mutant 20% versus WT
versus fludarabine plus 83 (all arms)a 27% (all arms)a,b 59% (all arms)a,b
cyclophosphamide (frontline)74
Chlorambucil versus 161 (9.9) Odds ratio for WT HR for WT versus HR for WT versus
chlorambucil plus rituximab versus mutant: 8.6 mutant: 1.8 (all arms) mutant: 3.8 (all arms)a
versus chlorambucil plus (all arms)a
obinutuzumab (frontline)190
Fludarabine plus 628 (11.5) • Mutant 52 versus WT • Mutant 12 versus WT • Mutant 30 versus WT
cyclophosphamide versus 92 (fludarabine plus 36 (fludarabine plus 90 (fludarabine plus
FCR (frontline)70 cyclophosphamide cyclophosphamide cyclophosphamide
arm)a arm)a arm)a
• Mutant 75 versus WT • Mutant 15 versus WT • Mutant 42 versus WT
98 (FCR arm)a 59 (FCR arm)a NR (FCR arm)a
Allo-​HSCT R/R142 72 (30) – HR for mutant versus HR for mutant versus
WT: 0.66 WT: 1.03
Targeted Ibrutinib (R/R)191 144 (100) Mutant 83 Mutant 63%c Mutant 75%c
Ibrutinib versus ofatumumab 154 (51) Mutant 91 versus WT Mutant 66% versus WT Mutant 86 versus WT
(R/R)145 (ibrutinib 92 (ibrutinib arm) 81% (ibrutinib arm)d 81 (ibrutinib arm)d
arm)
Idelalisib–rituximab versus 110 (46) – Mutant ~75% versus –
rituximab (R/R)135 WT 74% (idelalisib–
rituximab arm)e
Idelalisib–rituximab (frontline)154 64 (14) Mutant 100 versus Mutant 100% versus Mutant 100% versus
WT 96 WT 90%c WT 90%c
Venetoclax (R/R)138 107 (72) 79 (all patients) ~70% (all patients)e ~85% (all patients)e
ATM CIT Chlorambucil versus 161 (12.4) Odds ratio for WT HR for WT versus HR for WT versus
and/or chlorambucil plus versus mutant: 1.3 mutant: 1.5 (all arms) mutant: 0.8 (all arms)
del11q rituximab versus (all arms)
chlorambucil plus
obinutuzumab (frontline)190
Targeted Ibrutinib versus ofatumumab 154 (19) Mutant 93 versus Mutant 78% versus WT –
(R/R)145 WT 91 (ibrutinib arm) 73% (ibrutinib arm)d
Idelalisib–rituximab 160 (21) Mutant 85 versus – –
(frontline, R/R)192 WT 76
SF3B1 CIT Chlorambucil versus 437 (17) – Mutant 29 versus WT Mutant 54 versus
fludarabine versus fludarabine 39 (all arms)a WT 79 (all arms)a
plus cyclophosphamide
(frontline)193
Chlorambucil versus 161 (15.5) NS NS NS
chlorambucil plus
rituximab versus
chlorambucil plus
obinutuzumab (frontline)190
Fludarabine plus 628 (18) • Mutant 89 versus WT • Mutant 29 versus WT • Mutant 76
cyclophosphamide versus 88 (fludarabine plus 34 (fludarabine plus versus WT 86
FCR (frontline)70 cyclophosphamide cyclophosphamide (fludarabine plus
arm) arm)a cyclophosphamide
• Mutant 96 versus WT • Mutant 43 versus WT arm)
96 (FCR arm) 59 (FCR arm)a • Mutant NR versus
WT NR (FCR arm)
Allo-​HSCT Allo-​HSCT (R/R)142 72 (26) – HR for mutant versus HR for mutant versus
WT: 0.63 WT: 0.79
Targeted Ibrutinib versus ofatumumab 154 (31) Mutant 96 versus WT Mutant 65% versus –
(R/R)145 95 (ibrutinib arm) WT 79% (ibrutinib
arm)d

Nature Reviews | Clinical Oncology


Reviews

Table 3 (cont.) | Predictive value of genetic lesions according to treatment type.

genetic Treatment Study treatments (setting) number of ORR (%) mPFS (months) mOS (months)
lesion type patients
(% mutated)
NOTCH1 CIT Chlorambucil versus fludarabine 466 (10) – Mutant 22 versus WT Mutant 55 versus WT
versus fludarabine plus 27 (all arms)a 75 (all arms)a
cyclophosphamide (frontline)193
Chlorambucil versus 161 (20) NS NS NS
chlorambucil plus rituximab
versus chlorambucil plus
obinutuzumab (frontline)190
Fludarabine plus 628 (10) • Mutant 87 versus WT • Mutant 33 versus WT • Mutant 86 versus WT
cyclophosphamide versus 88 (fludarabine plus 34 (fludarabine plus 84 (fludarabine plus
FCR (frontline)70 cyclophosphamide cyclophosphamide cyclophosphamide
arm) arm) arm)
• Mutant 90 versus WT • Mutant 34 versus WT • Mutant 79 versus WT
96 (FCR arm) 57 (FCR arm)a NR (FCR arm)
Allo-​HSCT Allo-​HSCT (R/R)142 72 (14) – HR for mutant versus HR for mutant versus
WT: 0.69 WT: 0.68
Targeted Ibrutinib versus ofatumumab 154 (28) Mutant 93 versus WT Mutant 72% versus WT –
(R/R)145 91 (ibrutinib arm) 74% (ibrutinib arm)d
POT1 CIT Chlorambucil versus 161 (8.7) NS NS HR for WT versus
chlorambucil plus rituximab mutant: 2.9 (CIT arms)a
versus chlorambucil plus
obinutuzumab190 (frontline)
BIRC3 Targeted Ibrutinib versus ofatumumab 154 (14) Mutant 95 versus WT Mutant 81% versus WT –
(R/R)145 95 (ibrutinib arm) 72% (ibrutinib arm)d
Allo-​HSCT, allogeneic haematopoietic stem cell transplantation; CIT, chemoimmunotherapy ; FCR , fludarabine plus cyclophosphamide plus rituximab; HR , hazard
ratio; mOS, median overall survival; mPFS, median progression-​free survival; NR , not reached; NS: not significant; ORR , overall response rate; R/R , relapsed and/or
refractory ; WT, wild-​type. aStatistically significant; bAt 60 months; cAt 24 months; dAt 18 months; eAt 12 months.

of action, ibrutinib is active against CLL subtypes with PLCG2S707Y)147–149. BTKC481S mutations prevent the cova-
genetic lesions traditionally associated with resistance lent binding of ibrutinib to the BTK active site, resulting
to chemoimmunotherapy, namely TP53 or SF3B1 in a loss of inhibition of BTK enzymatic activity148,150.
lesions144 (Table 3). Post-​hoc analyses performed in the In addition, PLCG2 mutations are potentially gain-​of-
RESONATE trial, in which ibrutinib was compared with function lesions that promote BCR-​mediated signal-
ofatumumab in patients with relapsed and/or refractory ling independent of BTK inhibition148. Notably, the use
(R/R) CLL, showed that the overall response rate (ORR), of highly sensitive technologies (for example, droplet
PFS and OS in the ibrutinib arm were not influenced microfluid technologies) has enabled the detection of
by the presence of genetic lesions in ATM, NOTCH1, these mutations before the start of ibrutinib treatment147,
SF3B1 or BIRC3, although longer follow-​up is needed reinforcing the concept that external selection pressure
to conclusively validate these findings145. Conversely, the exerted by targeted therapies contributes to the selection
presence of genetic lesions in TP53 is correlated with of rare subclones that are already present before ther-
inferior ORR and PFS in patients with R/R CLL treated apy. Indeed, BTK and PLCG2 gene mutations have been
with ibrutinib, suggesting that, despite their superior described in ibrutinib-​treated patients with CLL up to
activity over chemoimmunotherapy in CLLs with TP53 15 months preceding the clinical relapse149,151, and they seem
lesions, current targeted therapies are not the final to occur with greater frequency in elderly patients with a
curative solution for CLL146. complex karyotype or TP53 disruptions149. Accordingly,
Sequential analysis of clonal dynamics in patients serial exome and transcriptome sequencing of patients
with CLL treated with ibrutinib did not reveal the with CLL treated with ibrutinib revealed the presence of
expansion of clones displaying common genetic driv- baseline mutations in pivotal downstream regulators
ers, for example SF3B1 or TP53 mutations45,147, which of BTK (for example, ITPKB and CDIPT) in one-​third of
is in contrast to the observation in patients treated with patients, which were associated with unfavourable PFS45.
chemoimmunotherapy41. Conversely, genetic analysis of In addition, mutations in ITPKB45, which have previ-
patients with CLL with disease progression during ibru- ously been described in lymphomas, and expansion
tinib treatment enabled the identification of potential of clones with deletions of 8p (del8p), which induces
mechanisms of resistance (Fig. 3). Ibrutinib binds cova- haploinsufficiency of the TRAIL receptor (TRAIL-​R), are
lently to the sulfhydryl group of C481 in the active site of genetic abnormalities that arise in patients who develop
BTK, resulting in the irreversible inhibition of its kinase resistance to ibrutinib45,147. Nevertheless, additional
activity. More than 80% of patients with acquired resist- experimental data, including analyses in animal models,
ance to ibrutinib harbour recurrent mutations involving are needed to determine the exact role of these mutations
the ibrutinib binding site of BTK (BTKC481S) or the BCR in the emergence of resistance to ibrutinib. A small frac-
signalling gene PLCG2 (PLCG2R665W, PLCG2L845F and tion of patients (~5%) receiving ibrutinib can develop a

www.nature.com/nrclinonc
Reviews

Active CLL the definition of a complex karyotype, the best method


to assess it, and its predictive influence with respect to
the targeted therapies remains to be established.
The BCR pathway has also been targeted by down-
TP53 wild type del17p and/or
TP53 mutation stream inhibition of PI3K. Idelalisib, a selective small-​
molecule inhibitor of PI3K catalytic subunit δ isoform
(PI3Kδ; also known as p110 δ isoform), which is speci­
IGHV-M, del13q IGHV-UM fically expressed in the haematopoietic compartment,
or trisomy 12 or del11q
has shown activity alone and in combination with rituxi­
mab in several B cell lymphoproliferative disorders135,154.
Although idelalisib is active in CLLs harbouring TP53
Fit Unfit Ibrutinib • Ibrutinib
• Ibrutinib • Ibrutinib • Venetoclax (if disruptions, data on the activity of this drug in the pres-
• FCR (BR >65 years) • Chlorambucil not candidate ence of other genetic lesions in CLL are limited (Table 3).
and CD20 mAb for ibrutinib) Along the same lines, little is known about the potential
molecular mechanisms of resistance to idelalisib.
In CLL, the BCL2 protein family members that regu-
R/R disease late the apoptotic process are skewed towards preventing
apoptosis for reasons that are not fully elucidated58,155,156.
Accordingly, blocking BCL2 protein function using the
BCL2 homology region 3 (BH3) mimetic venetoclax
R/R to CIT Intolerant of R/R to ibrutinib
ibrutinib leads to apoptosis in CLL cells and excellent oncologi­cal
control of the disease137. Venetoclax is highly active in
• Venetoclax patients with CLL with del17p and/or TP53 gene dys-
• Idelalisib + rituximab
• Clinical trial
regulation, which is consistent with its p53-independent
mechanism of action137,138. However, complex karyo­
types, but not TP53 disruption, predict progression
• Ibrutinib • Venetoclax + rituximab Cell therapy in patients treated with venetoclax121. A comparative
• Venetoclax + rituximab • Idelalisib + rituximab • Allo-HSCT
• Idelalisib + rituximab • CAR T cells genomic analysis of eight patients with CLL that was
resistant to or had progressed after venetoclax treatment,
Fig. 4 | Therapeutic algorithm for patients with Cll. Patients with chronic lymphocytic half of whom had a Richter syndrome transformation,
leukaemia (CLL) should be treated when they fulfil the International Workshop on Chronic showed acquisition of lesions in SF3B1 (subclonal), loss
Lymphocytic Leukemia (iwCLL) criteria8 for active disease. CLLs with TP53 disruptions of CDKN2A and/or CDKN2B, mutations in BRAF and
should be treated with ibrutinib or venetoclax (in patients who are not candidates for amplifications of CD274 (which encodes programmed
ibrutinib)158. In fit patients with low-​risk (that is, IGHV-​M status, del13q4 or trisomy 12) TP53 cell death 1 ligand 1 (PD-​L1))122. Functional studies in
wild-​type CLL , chemoimmunotherapy (CIT) combinations lead to similar outcomes to the same small panel of patients revealed that BRAF
ibrutinib, whereas the preferred treatment in unfit patients with low-​risk CLL is ibrutinib mutations, but not CDKN2A and/or CDKN2B deletions,
over combinations of chlorambucil and obinutuzumab or other anti-​CD20 antibodies
were associated with venetoclax resistance. Importantly,
(rituximab or ofatumumab)158,161. A general consensus exists on the use of front-​line ibrutinib
in patients with high-​risk CLL , even those without TP53 abnormalities. In patients with at the end of 2018, a study revealed that some patients
relapsed/refractory (R/R) disease after CIT, the most recommended options are ibrutinib or who are resistant to venetoclax acquire the hetero­
venetoclax plus rituximab, whereas idelalisib plus rituximab can be considered as the third zygous BCL2G101V mutation, which reduces sensitivity to
option133,135,145,158,167,184. Patients with R/R disease who are intolerant of ibrutinib can be venetoclax in vitro. The clinical influence of this muta-
salvaged with venetoclax plus rituximab or idelalisib plus rituximab. Finally , patients tion, usually detected at a low variant allele fraction,
progressing after ibrutinib can be treated with venetoclax plus rituximab or idelalisib plus requires confirmation157.
rituximab, or included in clinical trials, and subsequently considered for a cell therapy
depending on the characteristics of the patient and donor. Allo-​HSCT, allogeneic Influence on treatment strategies
haematopoietic stem cell transplantation; BR , bendamustine plus rituximab; During the past 5 years, the dissection of the genetic
CAR , chimeric antigen receptor ; FCR , fludarabine plus cyclophosphamide plus rituximab;
lesions underpinning CLL, along with the advent
mAb, monoclonal antibody.
of novel drugs, has shifted the treatment strategy in
patients with CLL from universal chemoimmunotherapy
Richter syndrome that is clonally related to the under- to a more individualized approach. At diagnosis, assess-
lying CLL, usually during the first 2 years of treatment ment of IGHV mutational status, molecular cytogenetics
and sometimes with bone marrow involvement120 (Fig. 3). (using FISH) and mutational analysis of the TP53 gene
These transformations typically exhibit MYC gains and are currently recommended in the iwCLL guidelines to
lack the common BTK and PLCG2 mutations. ascertain the prognostic risk in a given patient105. In a
The presence of complex karyotypes has been asso- patient requiring treatment, these molecular tests are
ciated with inferior responses to both chemoimmuno­ mandatory because they help in identifying the best
therapy and ibrutinib 149, suggesting that genomic therapeutic strategy (Fig. 4). In patients included in clini­
instability has an unfavourable effect on response to cal trials, the additional analysis of biomarkers directly
treatment. Data from 2018 suggest that the adverse influ- informing the response to targeted therapy is advisable.
ence of complex karyotypes in half the patients treated Finally, complex karyotype analysis, although poten-
with ibrutinib is independent of the presence of TP53 tially useful in predicting response, is still considered
genetic abnormalities152,153. Nonetheless, a consensus on investigational.

Nature Reviews | Clinical Oncology


Reviews

First-​line treatment algorithm with low-​risk CLL will still be a subject of debate until
A general consensus exists that treatment-​naive patients mature results from ongoing randomized clinical trials
requiring therapy should be tested for the presence of comparing both treatments provide conclusive data on
del17p and TP53 disruptions (Fig. 4). If these lesions this issue.
are present, even at the subclonal level75, these patients
should not be treated with genotoxic drugs (alkylating Treatment of relapsed/refractory CLL
agents, anthracyclines or purine analogues), given that Salvage chemoimmunotherapy in patients with R/R CLL
genetic lesions hampering DNA repair predict a poor who have previously been treated with chemotherapy
response38,75,82. Indeed, the median PFS of patients with leads to short-​lasting responses, with a median PFS of
CLL harbouring TP53 lesions treated using FCR or less than 2 years165,166. This suboptimal response is mainly
other chemoimmunotherapy combinations is less than due to the acquisition of resistant clones selected by pre-
18 months71. Thus, these patients are candidates for ther- vious therapies (Fig. 3) and to difficulties in completing
apies with mechanisms of action not requiring the pres- the planned treatment owing to their excessive toxicity.
ence of an intact and functional DNA repair machinery. In the setting of R/R CLL after chemoimmunotherapy,
Ibrutinib is the most recommended upfront treatment in a clear indication exists for the use of targeted therapies
patients with TP53 mutations158, leading to an estimated (Fig. 4). The median PFS in heavily pretreated patients
median PFS of more than 30 months152. In patients who receiving salvage therapy with ibrutinib is 51 months146.
are not eligible to receive ibrutinib (for example, owing The combination of venetoclax plus rituximab was
to specific comorbidities, drug interactions or patient approved in 2018 by the FDA for this indication, and
choice), venetoclax is an excellent alternative138. is an interesting early option for the treatment of R/R
In patients without TP53 disruptions, assessment of CLL167. Approximately 15% of patients in the frontline
the IGHV mutational status and physical fitness are key and the R/R settings develop resistance to ibrutinib
to individualized treatment. Long-​term follow-up data (parti­cularly those with TP53 disruptions), which is
from patients with CLL treated with FCR, the mainstay probably attributable to the aforementioned molecular
of chemoimmunotherapy in CLL, show that approxi- mechanisms of resistance to ibrutinib. Around 65% of
mately half of the patients with IGHV-​M CLL have a these patients respond to venetoclax168, but they should
durable response of more than 10 years128,130. Thus, FCR be further considered for allogeneic HSCT, chimeric
or bendamustine plus rituximab (BR) have been the pre- antigen receptor (CAR) T cell therapy or participation
ferred chemoimmunotherapy regimens for fit patients in clinical trials, given that the median time to progres-
with IGHV-​M disease, whereas less intense combina- sion of patients failing to ibrutinib and treated with
tions of chlorambucil with a CD20 monoclonal antibody venetoclax is ~25 months.
(such as obinutuzumab, ofatumumab or rituximab) can Several additional BTK inhibitors are currently
be recommended for those patients who will probably being tested, alone and in combination, in the front-
not tolerate FCR or BR128–130,159. However, data published line and the R/R settings (Fig. 2). Acalabrutinib (ACP-
in 2018 and 2019 from three indepen­dent randomized 196), which was approved for treatment of mantle cell
clinical trials showed that, in low-​risk patients with lymphoma in 2018, seems to have a better toxicity pro-
CLL, ibrutinib with or without monoclonal anti-​CD20 file than ibrutinib owing to its greater specificity and,
antibodies leads to PFS outcomes similar to those of therefore, fewer off-​target effects. Despite the fact that
chemoimmunotherapy160–162Although additional follow-​ acalabrutinib results in an ORR of 95% in patients with
up monitoring is still required, these data indicate that R/R disease169, whether this agent is more active than
ibrutinib should be placed at the same level as chemo­ ibrutinib requires a longer follow-​up and the results
immunotherapy in the therapeutic algorithm. Notably, in of a phase III study comparing acalabrutinib versus
the clinically unfit patients with TP53 wild-​type disease, ibrutinib (NCT02477696). Vecabrutinib (SNS-062) is
ibrutinib is usually the preferred option owing to both another BCR inhibitor that seems to be active in vitro
its high activity leading to an estimated PFS of 75% at in CLL cells with the common BTKC481S resistance muta-
4 years, and its good safety profile145,158,163. By contrast, tion170 and in a preliminary study using a CLL adop-
patients with IGHV-​UM CLL have a PFS of 33% at tive transfer model171. Whether or not this will translate
5 years when treated with chemoimmuno­therapy130. For into adequate clinical activity is currently being eval-
this high-​risk population, a growing consensus exists on uated in a clinical trial (NCT03037645)172. Other BTK
the use of ibrutinib in the front-​line setting, regardless of inhibitors (BGB-3111 (NCT03336333), CT-1530
age and comorbidities (reviewed in164). This consensus (NCT02981745), tirabrutinib (NCT03740529), M7583
is based on the observation that IGHV mutational status (NCT02825836), DTRMWXHS (NCT02891590) and
does not influence PFS in patients treated with ibruti- CC-292 (NCT01744626)) and SYK inhibitors (entos-
nib146, which was confirmed in 2019 by the results of the pletinib (NCT02983617), TAK-659 (NCT02954406)
aforementioned clinical trials160–162. and cerdulatinib (NCT01994382)) are currently under
An important aspect of BTK inhibitor therapy is that investigation in R/R CLL (Fig. 2).
it should be administered continuously because minimal Idelalisib, an inhibitor of PI3Kδ, has also been
residual disease (MRD)-negative responses cannot be approved in combination with rituximab for the
achieved with these agents145. This characteristic trans- treatment of CLL135. Unfortunately, PI3Kδ inhibition
lates into several issues relating to long-​term toxicities, has been associated with a particular toxicity profile
compliance, drug interactions and cost. Thus, the use characterized by autoimmune complications and an
of ibrutinib before chemoimmunotherapy in patients increased risk of opportunistic infections, particularly

www.nature.com/nrclinonc
Reviews

in treatment-​naive patients (diarrhoea 42%, pneumo- translate into deeper responses and shorter treatment
nitis 20% and hepatotoxicity 16%)173. Thus, the use of schedules than those associated with continuous admin-
idelalisib is usually restricted to patients with R/R dis- istration of ibrutinib (reviewed in179). Preliminary results
ease after treatment with ibrutinib or venetoclax. In addi- with the combinations of ibrutinib and venetoclax,
tion, duvelisib was approved at the end of 2018 for the venetoclax and obinutuzumab or the triplet bendamus-
treatment of R/R CLL based on a randomized clinical tine, obinutuzumab and venetoclax showed ORRs of
trial (NTC02004522) comparison of this agent against >90%, including a substantial number of patients with
ofatumumab174. Other PI3K inhibitors, some of which undetectable MRD responses, with a low toxicity pro-
have toxicity profiles distinct from that of idelalisib, file in patients with R/R CLL180–183. Nonetheless, iden-
are currently under investigation in clinical trials in tification of the long-​term activity or toxicity as well as
patients with CLL and other lymphoproliferative dis­ the best way to combine or sequence these treatments
orders in the R/R setting (umbralisib (NCT02268851) await the results of the numerous ongoing clinical trials
and YY-20394 (NCT03757000)). Finally, other com- (NCT02756897, NCT03128879 and NCT02950051).
pounds targeting specific pathways, including NOTCH1
signalling (CB-103 (NCT03422679), brontictuzumab Conclusions
(NCT01703572), RO4929097 (NCT01158274), During the past 5 years, great progress has been made
LY3039478 (NCT01695005) and Pf-03084014 in the clinical application of new genomic methods for
(NCT00878189)), inhibition of nuclear export of proteins dissecting the genetic aetiology of CLL as well as in the
(selinexor (NCT02303392), RNA processing (SL-801 identification of patterns of clonal evolution, clinical
(NCT02667873)), inflammatory pathways (IMO-8400 progression and therapeutic resistance. Accordingly,
(NCT02363439) and CA-4948 (NCT03328078)), MCL-1 new prognostic and predictive biomarkers and novel
inhibitors (MIK665 (NCT02992483) and AMG176 targeted therapies have been developed, which are
(NCT03797261)) and the DNA damage response being translated into improved risk stratification tools,
pathway (AZD6738 (NCT01955668), BAY1895344 prolonged PFS and, ultimately, extended OS in patients
(NCT03188965), APR-246 (NCT00900614) and COTI-2 with CLL. However, this progress has opened the door
(NCT02433626)) are currently under investigation in to new challenges in several major areas. Clear evi-
early phase clinical trials (Fig. 2). dence is emerging that most genetic events leading to
No standard treatment strategy exists for patients tumour progression or drug resistance are present in
with Richter syndrome. Most of these patients have very early pretreatment stages of the disease, and that
TP53 disruptions, which preclude the response to the their stochastic accumulation is positively correlated
rituximab–cyclophosphamide, doxorubicin, vincristine with the number of tumour cells and their divisions.
and prednisone (R-​CHOP)-like chemotherapy regimens Thus, future studies should explore earlier therapeutic
that are usually used in patients with high-​grade lym- interventions at diagnosis or even during clinical remis-
phoma (reviewed in114). Of note, interesting data have sions in patients with high-​risk disease, when the hetero­
been obtained with the combination of ibrutinib or other geneity and abundance of different subclones are still
BCR inhibitors and immune-​checkpoint inhibitors175,176. generally low. Furthermore, the responses to targeted
Of patients with Richter transformation who were pre- therapies, for instance to ibrutinib and venetoclax, are
viously refractory to chemoimmunotherapy, 65% had very encouraging, yet the determinants of response or
a response to treatment with ibrutinib plus nivolumab. non-​response are only partially understood. Thus, the
Unfortunately, responses to chemotherapy or immune-​ coupling of each clinical trial with extensive, prospective
checkpoint inhibitors are generally less than 12 months. searches for biomarkers of response is needed. Finally,
Thus, patients with Richter transformation usually the enormous and growing costs of the new drugs pose
undergo consolidation cell therapy with allogeneic formidable socioeconomic challenges that go far beyond
HSCT or CAR T cell therapy177,178. any strictly biomedical considerations.
Novel targeted therapies have been combined with
the aim of achieving synergistic activities that could Published online xx xx xxxx

1. Swerdlow, S. H. et al. The 2016 revision of the World 6. Goldin, L. R. et al. Whole exome sequencing in 11. Rai, K. R. et al. Clinical staging of chronic lymphocytic
Health Organization classification of lymphoid families with CLL detects a variant in Integrin β 2 leukemia. Blood 46, 219–234 (1975).
neoplasms. Blood 127, 2375–2390 (2016). associated with disease susceptibility. Blood 128, 12. Binet, J. L. et al. A new prognostic classification
2. National Cancer Institute. Cancer stat facts: 2261–2263 (2016). of chronic lymphocytic leukemia derived from a
leukemia — chronic lymphocytic leukemia (CLL). 7. Law, P. J. et al. Genome-​wide association analysis multivariate survival analysis. Cancer 48, 198–206
SEER https://seer.cancer.gov/statfacts/html/clyl.html implicates dysregulation of immunity genes in chronic (1981).
(2019). lymphocytic leukaemia. Nat. Commun. 8, 14175 (2017). 13. Rai, K. R. Progress in chronic lymphocytic leukaemia:
3. Wu, S.-J. et al. The incidence of chronic lymphocytic 8. Abrisqueta, P. et al. Improving survival in patients with a historical perspective. Baillières Clin. Haematol. 6,
leukemia in Taiwan, 1986-2005: a distinct increasing chronic lymphocytic leukemia (1980–2008): the 757–765 (1993).
trend with birth-​cohort effect. Blood 116, 4430–4435 Hospital Clinic of Barcelona experience. Blood 114, 14. Crespo, M. et al. ZAP-70 expression as a surrogate for
(2010). 2044–2050 (2009). immunoglobulin-​variable-region mutations in chronic
4. Wu, S.-J., Chiang, C.-J., Lin, C.-T., Tien, H.-F. & Lai, M.-S. 9. Hallek, M. et al. Guidelines for the diagnosis and lymphocytic leukemia. N. Engl. J. Med. 348,
A nationwide population-​based cross-​sectional treatment of chronic lymphocytic leukemia: a report 1764–1775 (2003).
comparison of hematological malignancies incidences from the International Workshop on Chronic 15. Rossi, D. et al. Integrated mutational and cytogenetic
between Taiwan and the United States of America. Lymphocytic Leukemia updating the National Cancer analysis identifies new prognostic subgroups in chronic
Ann. Hematol. 95, 165–167 (2016). Institute–Working Group 1996 guidelines. Blood 111, lymphocytic leukemia. Blood 121, 1403–1412 (2013).
5. Goldin, L. R., Pfeiffer, R. M., Li, X. & Hemminki, K. 5446–5456 (2008). 16. Cramer, P. & Hallek, M. Prognostic factors in chronic
Familial risk of lymphoproliferative tumors in families 10. Campo, E., Harris, N. L., Jaffe, E. S., Stein, H. lymphocytic leukemia—what do we need to know?
of patients with chronic lymphocytic leukemia: results & Thiele, J. WHO Classification of Tumours Nat. Rev. Clin. Oncol. 8, 38 (2011).
from the Swedish Family-​Cancer Database. Blood 104, of Haematopoietic and Lymphoid Tissues 17. International CLL-​IPI working group. An international
1850–1854 (2004). (eds Swerdlow, S. H. et al.) (IARC, 2017). prognostic index for patients with chronic lymphocytic

Nature Reviews | Clinical Oncology


Reviews

leukaemia (CLL-​IPI): a meta-​analysis of individual of recurrent RPS15 mutations. Blood 127, 71. Zenz, T. et al. Monoallelic TP53 inactivation
patient data. Lancet Oncol. 17, 779–790 (2016). 1007–1016 (2016). is associated with poor prognosis in chronic
18. Burger, J. A. & Wiestner, A. Targeting B cell receptor 44. Kasar, S. et al. Whole-​genome sequencing reveals lymphocytic leukemia: results from a detailed genetic
signalling in cancer: preclinical and clinical advances. activation-​induced cytidine deaminase signatures characterization with long-​term follow-​up. Blood 112,
Nat. Rev. Cancer 18, 148–167 (2018). during indolent chronic lymphocytic leukaemia 3322–3329 (2008).
19. Caligaris-​Cappio, F., Gobbi, M., Bofill, M. & Janossy, G. evolution. Nat. Commun. 6, 8866 (2015). 72. Zenz, T. et al. TP53 mutation and survival in chronic
Infrequent normal B lymphocytes express features of 45. Landau, D. A. et al. The evolutionary landscape of lymphocytic leukemia. J. Clin. Oncol. 28, 4473–4479
B-​chronic lymphocytic leukemia. J. Exp. Med. 155, chronic lymphocytic leukemia treated with ibrutinib (2010).
623–628 (1982). targeted therapy. Nat. Commun. 8, 2185 (2017). 73. Döhner, H. et al. p53 gene deletion predicts for poor
20. Stall, A. M. et al. Ly-1 B cell clones similar to human 46. Vogelstein, B. et al. Cancer genome landscapes. survival and non-​response to therapy with purine
chronic lymphocytic leukemias routinely develop in Science 339, 1546–1558 (2013). analogs in chronic B cell leukemias. Blood 85,
older normal mice and young autoimmune (New 47. Alexandrov, L. B. et al. Signatures of mutational 1580–1589 (1995).
Zealand Black-​related) animals. Proc. Natl Acad. Sci. processes in human cancer. Nature 500, 415–421 74. Gonzalez, D. et al. Mutational status of the TP53 gene
U. S. A. 85, 7312–7316 (1988). (2013). as a predictor of response and survival in patients with
21. Klein, U. et al. Gene expression profiling of B cell 48. Fabbri, G. & Dalla-​Favera, R. The molecular chronic lymphocytic leukemia: results from the LRF
chronic lymphocytic leukemia reveals a homogeneous pathogenesis of chronic lymphocytic leukaemia. CLL4 trial. J. Clin. Oncol. 29, 2223–2229 (2011).
phenotype related to memory B cells. J. Exp. Med. Nat. Rev. Cancer 16, 145–162 (2016). 75. Rossi, D. et al. Clinical impact of small TP53 mutated
194, 1625–1638 (2001). 49. Rosenquist, R., Beà, S., Du, M.-Q., Nadel, B. subclones in chronic lymphocytic leukemia. Blood
22. Rosenwald, A. et al. Relation of gene expression & Pan-Hammarström, Q. Genetic landscape and 123, 2139–2147 (2014).
phenotype to immunoglobulin mutation genotype deregulated pathways in B cell lymphoid malignancies. 76. Trbusek, M. et al. Missense mutations located in
in B cell chronic lymphocytic leukemia. J. Exp. Med. J. Intern. Med. 282, 371–394 (2017). structural p53 DNA-​binding motifs are associated
194, 1639–1648 (2001). 50. Döhner, H. et al. Genomic aberrations and survival in with extremely poor survival in chronic lymphocytic
23. Seifert, M. et al. Cellular origin and pathophysiology chronic lymphocytic leukemia. N. Engl. J. Med. 343, leukemia. J. Clin. Oncol. 29, 2703–2708 (2011).
of chronic lymphocytic leukemia. J. Exp. Med. 209, 1910–1916 (2000). 77. Yu, L. et al. Survival of Del17p CLL depends on
2183–2198 (2012). 51. Klein, U. et al. The DLEU2/miR-15a/16-1 cluster genomic complexity and somatic mutation.
24. Kulis, M. et al. Epigenomic analysis detects widespread controls B cell proliferation and its deletion leads to Clin. Cancer Res. 23, 735–745 (2017).
gene-​body DNA hypomethylation in chronic lymphocytic chronic lymphocytic leukemia. Cancer Cell 17, 28–40 78. Austen, B. et al. Mutations in the ATM gene lead to
leukemia. Nat. Genet. 44, 1236–1242 (2012). (2010). impaired overall and treatment-​free survival that is
25. Agathangelidis, A. et al. Stereotyped B cell receptors 52. Rawstron, A. C. Monoclonal B cell lymphocytosis and independent of IGVH mutation status in patients with
in one-​third of chronic lymphocytic leukemia: a chronic lymphocytic leukemia. N. Engl. J. Med. 359, B-​CLL. Blood 106, 3175–3182 (2005).
molecular classification with implications for targeted 575–583 (2008). 79. Rossi, D. et al. Disruption of BIRC3 associates with
therapies. Blood 119, 4467–4475 (2012). 53. Migliazza, A. et al. Nucleotide sequence, transcription fludarabine chemorefractoriness in TP53 wild-​type
26. Vardi, A. et al. Immunogenetic studies of chronic map, and mutation analysis of the 13q14 chromosomal chronic lymphocytic leukemia. Blood 119, 2854–2862
lymphocytic leukemia: revelations and speculations region deleted in B cell chronic lymphocytic leukemia. (2012).
about ontogeny and clinical evolution. Cancer Res. Blood 97, 2098–2104 (2001). 80. Stankovic, T. & Skowronska, A. The role of ATM
74, 4211–4216 (2014). 54. Kalachikov, S. et al. Cloning and gene mapping of the mutations and 11q deletions in disease progression in
27. Tobin, G., Thunberg, U. & Johnson, A. Chronic chromosome 13q14 region deleted in chronic chronic lymphocytic leukemia. Leuk. Lymphoma 55,
lymphocytic leukemias utilizing the VH3-21 gene lymphocytic leukemia. Genomics 42, 369–377 (1997). 1227–1239 (2014).
display highly restricted Vlambda2-14 gene use and 55. Calin, G. A. et al. Frequent deletions and down-​ 81. Skowronska, A. et al. Biallelic ATM inactivation
homologous CDR3s: implicating recognition of a regulation of micro- RNA genes miR15 and miR16 at significantly reduces survival in patients treated on
common antigen epitope. Blood 101, 4952–4957 13q14 in chronic lymphocytic leukemia. Proc. Natl the United Kingdom Leukemia Research Fund Chronic
(2003). Acad. Sci. U. S. A. 99, 15524–15529 (2002). Lymphocytic Leukemia 4 trial. J. Clin. Oncol. 30,
28. Stamatopoulos, K., Agathangelidis, A., Rosenquist, R. 56. Palamarchuk, A. et al. 13q14 deletions in CLL 4524–4532 (2012).
& Ghia, P. Antigen receptor stereotypy in chronic involve cooperating tumor suppressors. Blood 115, 82. Nadeu, F. et al. Clinical impact of clonal and subclonal
lymphocytic leukemia. Leukemia 31, 282–291 (2017). 3916–3922 (2010). TP53, SF3B1, BIRC3, NOTCH1, and ATM mutations
29. Minden, M. D. et al. Chronic lymphocytic leukaemia is 57. Hammarsund, M. et al. Characterization of a novel in chronic lymphocytic leukemia. Blood 127,
driven by antigen-​independent cell-​autonomous B-CLL candidate gene — DLEU7 — located in the 2122–2130 (2016).
signalling. Nature 489, 309–312 (2012). 13q14 tumor suppressor locus. FEBS Lett. 556, 83. Ramsay, A. J. et al. POT1 mutations cause telomere
30. Husby, S. & Grønbæk, K. Mature lymphoid 75–80 (2004). dysfunction in chronic lymphocytic leukemia.
malignancies: origin, stem cells, and chronicity. 58. Cimmino, A. et al. miR-15 and miR-16 induce Nat. Genet. 45, 526–530 (2013).
Blood Adv. 1, 2444–2455 (2017). apoptosis by targeting BCL2. Proc. Natl Acad. Sci. 84. Rossi, D. et al. Mutations of the SF3B1 splicing factor
31. Kikushige, Y. et al. Self-​renewing hematopoietic stem U. S. A. 102, 13944–13949 (2005). in chronic lymphocytic leukemia: association with
cell is the primary target in pathogenesis of human 59. Belver, L. & Ferrando, A. The genetics and mechanisms progression and fludarabine-​refractoriness. Blood
chronic lymphocytic leukemia. Cancer Cell 20, of T cell acute lymphoblastic leukaemia. Nat. Rev. 118, 6904–6908 (2011).
246–259 (2011). Cancer 16, 494–507 (2016). 85. Wang, L. et al. Transcriptomic characterization of
32. Damm, F. et al. Acquired initiating mutations in early 60. Guruharsha, K. G., Kankel, M. W. & Artavanis-​ SF3B1 mutation reveals its pleiotropic effects in
hematopoietic cells of CLL patients. Cancer Discov. 4, Tsakonas, S. The Notch signalling system: recent chronic lymphocytic leukemia. Cancer Cell 30,
1088–1101 (2014). insights into the complexity of a conserved pathway. 750–763 (2016).
33. Marsilio, S. et al. Somatic CLL mutations occur at Nat. Rev. Genet. 13, 654–666 (2012). 86. Mansouri, L. et al. Functional loss of IκBε leads to
multiple distinct hematopoietic maturation stages: 61. Arruga, F. et al. Functional impact of NOTCH1 NF-κB deregulation in aggressive chronic lymphocytic
documentation and cautionary note regarding cell mutations in chronic lymphocytic leukemia. Leukemia leukemia. J. Exp. Med. 212, 833–843 (2015).
fraction purity. Leukemia 32, 1041–1044 (2018). 28, 1060–1070 (2014). 87. Martínez-​Trillos, A. et al. Mutations in TLR/MYD88
34. Jaiswal, S. et al. Age-​related clonal hematopoiesis 62. Fabbri, G. et al. Common nonmutational NOTCH1 pathway identify a subset of young chronic lymphocytic
associated with adverse outcomes. N. Engl. J. Med. activation in chronic lymphocytic leukemia. Proc. Natl leukemia patients with favorable outcome. Blood 123,
371, 2488–2498 (2014). Acad. Sci. U. S. A. 114, E2911–E2919 (2017). 3790–3796 (2014).
35. Genovese, G. et al. Clonal hematopoiesis and blood-​ 63. Rossi, D. et al. Mutations of NOTCH1 are an 88. Martínez-​Trillos, A. et al. Clinical impact of MYD88
cancer risk inferred from blood DNA sequence. independent predictor of survival in chronic mutations in chronic lymphocytic leukemia. Blood
N. Engl. J. Med. 371, 2477–2487 (2014). lymphocytic leukemia. Blood 119, 521–529 (2012). 127, 1611–1613 (2016).
36. Fabbri, G. et al. Analysis of the chronic lymphocytic 64. Baliakas, P. et al. Recurrent mutations refine prognosis 89. Qin, S.-C. et al. MYD88 mutations predict unfavorable
leukemia coding genome: role of NOTCH1 mutational in chronic lymphocytic leukemia. Leukemia 29, prognosis in chronic lymphocytic leukemia patients
activation. J. Exp. Med. 208, 1389–1401 (2011). 329–336 (2015). with mutated IGHV gene. Blood Cancer J. 7, 651
37. Wang, L. et al. SF3B1 and other novel cancer genes in 65. Fabbri, G. et al. Genetic lesions associated with chronic (2017).
chronic lymphocytic leukemia. N. Engl. J. Med. 365, lymphocytic leukemia transformation to Richter 90. Santos, F. P. S. & O’Brien, S. Small lymphocytic
2497–2506 (2011). syndrome. J. Exp. Med. 210, 2273–2288 (2013). lymphoma and chronic lymphocytic leukemia: are they
38. Landau, D. A. et al. Evolution and impact of subclonal 66. Rossi, D. et al. The genetics of Richter syndrome the same disease? Cancer J. 18, 396–403 (2012).
mutations in chronic lymphocytic leukemia. Cell 152, reveals disease heterogeneity and predicts survival 91. Del Giudice, I. et al. NOTCH1 mutations in +12 chronic
714–726 (2013). after transformation. Blood 117, 3391–3401 lymphocytic leukemia (CLL) confer an unfavorable
39. Puente, X. S. et al. Whole-​genome sequencing (2011). prognosis, induce a distinctive transcriptional profiling
identifies recurrent mutations in chronic lymphocytic 67. Balatti, V. et al. NOTCH1 mutations in CLL associated and refine the intermediate prognosis of +12 CLL.
leukaemia. Nature 475, 101–105 (2011). with trisomy 12. Blood 119, 329–331 (2012). Haematologica 97, 437–441 (2012).
40. Puente, X. S. et al. Non-​coding recurrent mutations 68. Benedetti, D. et al. NOTCH1 mutations are associated 92. Chigrinova, E. et al. Two main genetic pathways lead to
in chronic lymphocytic leukaemia. Nature 526, with high CD49d expression in chronic lymphocytic the transformation of chronic lymphocytic leukemia
519–524 (2015). leukemia: link between the NOTCH1 and the NF-​κB to Richter syndrome. Blood 122, 2673–2682 (2013).
41. Landau, D. A. et al. Mutations driving CLL and their pathways. Leukemia 32, 654–662 (2017). 93. Landau, D. A. et al. Locally disordered methylation
evolution in progression and relapse. Nature 526, 69. Pozzo, F. et al. NOTCH1-mutated chronic lymphocytic forms the basis of intratumor methylome variation in
525–530 (2015). leukemia cells are characterized by a MYC-​related chronic lymphocytic leukemia. Cancer Cell 26,
42. Quesada, V. et al. Exome sequencing identifies overexpression of nucleophosmin 1 and ribosome-​ 813–825 (2014).
recurrent mutations of the splicing factor SF3B1 associated components. Leukemia 31, 2407–2415 94. Beekman, R. et al. The reference epigenome and
gene in chronic lymphocytic leukemia. Nat. Genet. (2017). regulatory chromatin landscape of chronic lymphocytic
44, 47–52 (2012). 70. Stilgenbauer, S. et al. Gene mutations and treatment leukemia. Nat. Med. 24, 868–880 (2018).
43. Ljungström, V. et al. Whole-​exome sequencing in outcome in chronic lymphocytic leukemia: results from 95. Timp, W. & Feinberg, A. P. Cancer as a dysregulated
relapsing chronic lymphocytic leukemia: clinical impact the CLL8 trial. Blood 123, 3247–3254 (2014). epigenome allowing cellular growth advantage at the

www.nature.com/nrclinonc
Reviews

expense of the host. Nat. Rev. Cancer 13, 497–510 ibrutinib treatment for relapsed chronic lymphocytic a phase 2, single-​arm trial. Lancet Oncol. 16,
(2013). leukemia. Hematol. Oncol. 36, 600–603 (2018). 169–176 (2015).
96. Gaiti, F. et al. Epigenetic evolution and lineage 121. Anderson, M. A. et al. Clinico-​pathological features 145. Brown, J. R. et al. Extended follow-​up and impact
histories of chronic lymphocytic leukaemia. Nature and outcomes of progression of CLL on the BCL2 of high-​risk prognostic factors from the phase 3
569, 576–580 (2019). inhibitor venetoclax. Blood 129, 3362–3370 (2017). RESONATE study in patients with previously treated
97. World Health Organization International Agency for 122. Herling, C. D. et al. Clonal dynamics towards the CLL/SLL. Leukemia 32, 83–91 (2018).
Research on Cancer. WHO Classification of Tumours development of venetoclax resistance in chronic 146. O’Brien, S. et al. Single-​agent ibrutinib in treatment-​
of Haematopoietic and Lymphoid Tissues 4th edn lymphocytic leukemia. Nat. Commun. 9, 727 (2018). naïve and relapsed/refractory chronic lymphocytic
(eds Swerdlow, S. H. et al.) (IARC, 2008). 123. Hamblin, T. J., Davis, Z., Gardiner, A., Oscier, D. G. leukemia: a 5-year experience. Blood 131, 1910–1919
98. Nieto, W. G. et al. Increased frequency (12%) of & Stevenson, F. K. Unmutated Ig VH genes are (2018).
circulating chronic lymphocytic leukemia-​like B cell associated with a more aggressive form of chronic 147. Burger, J. A. et al. Clonal evolution in patients with
clones in healthy subjects using a highly sensitive lymphocytic leukemia. Blood 94, 1848–1854 (1999). chronic lymphocytic leukaemia developing resistance
multicolor flow cytometry approach. Blood 114, 124. Damle, R. N. et al. Ig V gene mutation status and to BTK inhibition. Nat. Commun. 7, 11589 (2016).
33–37 (2009). CD38 expression as novel prognostic indicators in 148. Woyach, J. A. et al. Resistance mechanisms for the
99. Almeida, J. et al. CLL-​like B-​lymphocytes are chronic lymphocytic leukemia. Blood 94, 1840–1847 Bruton’s tyrosine kinase inhibitor ibrutinib. N. Engl.
systematically present at very low numbers in (1999). J. Med. 370, 2286–2294 (2014).
peripheral blood of healthy adults. Leukemia 25, 125. Jeromin, S. et al. SF3B1 mutations correlated to 149. Woyach, J. A. et al. BTKC481S-​mediated resistance to
718–722 (2011). cytogenetics and mutations in NOTCH1, FBXW7, ibrutinib in chronic lymphocytic leukemia. J. Clin. Oncol.
100. Strati, P. & Shanafelt, T. D. Monoclonal B cell MYD88, XPO1 and TP53 in 1160 untreated CLL 35, 1437–1443 (2017).
lymphocytosis and early-​stage chronic lymphocytic patients. Leukemia 28, 108–117 (2014). 150. Furman, R. R. et al. Ibrutinib resistance in chronic
leukemia: diagnosis, natural history, and risk 126. Nadeu, F. et al. Clinical impact of the subclonal lymphocytic leukemia. N. Engl. J. Med. 370,
stratification. Blood 126, 454–462 (2015). architecture and mutational complexity in chronic 2352–2354 (2014).
101. Shanafelt, T. D., Ghia, P., Lanasa, M. C., Landgren, O. lymphocytic leukemia. Leukemia 32, 645–653 (2017). 151. Ahn, I. E. et al. Clonal evolution leading to ibrutinib
& Rawstron, A. C. Monoclonal B cell lymphocytosis 127. Keating, M. J. et al. Early results of a resistance in chronic lymphocytic leukemia. Blood
(MBL): biology, natural history and clinical chemoimmunotherapy regimen of fludarabine, 129, 1469–1479 (2017).
management. Leukemia 24, 512–520 (2010). cyclophosphamide, and rituximab as initial therapy 152. Jones, J. et al. Evaluation of 230 patients with
102. Landgren, O. et al. B cell clones as early markers for for chronic lymphocytic leukemia. J. Clin. Oncol. 23, relapsed/refractory deletion 17p chronic lymphocytic
chronic lymphocytic leukemia. N. Engl. J. Med. 360, 4079–4088 (2005). leukaemia treated with ibrutinib from 3 clinical trials.
659–667 (2009). 128. Thompson, P. A. et al. Fludarabine, cyclophosphamide, Br. J. Haematol. 182, 504–512 (2018).
103. Vardi, A. et al. Immunogenetics shows that not all and rituximab treatment achieves long-​term disease-​ 153. Rigolin, G. M. et al. In chronic lymphocytic leukaemia
MBL are equal: the larger the clone, the more similar free survival in IGHV-​mutated chronic lymphocytic with complex karyotype, major structural
to CLL. Blood 121, 4521–4528 (2013). leukemia. Blood 127, 303–309 (2016). abnormalities identify a subset of patients with
104. Ghia, P., Melchers, F. & Rolink, A. G. Age-​dependent 129. Hallek, M. et al. Addition of rituximab to fludarabine inferior outcome and distinct biological characteristics.
changes in B lymphocyte development in man and and cyclophosphamide in patients with chronic Br. J. Haematol. 181, 229–233 (2018).
mouse. Exp. Gerontol. 35, 159–165 (2000). lymphocytic leukaemia: a randomised, open-​label, 154. O’Brien, S. M. et al. A phase 2 study of idelalisib plus
105. Hallek, M. et al. iwCLL guidelines for diagnosis, phase 3 trial. Lancet 376, 1164–1174 (2010). rituximab in treatment-​naive older patients with chronic
indications for treatment, response assessment, 130. Fischer, K. et al. Long-​term remissions after FCR lymphocytic leukemia. Blood 126, 2686–2694 (2015).
and supportive management of CLL. Blood 131, chemoimmunotherapy in previously untreated 155. Del Gaizo Moore, V. et al. Chronic lymphocytic
2745–2760 (2018). patients with CLL: updated results of the CLL8 trial. leukemia requires BCL2 to sequester prodeath BIM,
106. Dagklis, A. et al. The immunoglobulin gene repertoire Blood 127, 208–215 (2016). explaining sensitivity to BCL2 antagonist ABT-737.
of low-​count chronic lymphocytic leukemia (CLL)-like 131. Bosch, F. et al. Rituximab, fludarabine, J. Clin. Invest. 117, 112–121 (2007).
monoclonal B lymphocytosis is different from CLL: cyclophosphamide, and mitoxantrone: a new, highly 156. van Delft, M. F. et al. The BH3 mimetic ABT-737
diagnostic implications for clinical monitoring. Blood active chemoimmunotherapy regimen for chronic targets selective Bcl-2 proteins and efficiently induces
114, 26–32 (2009). lymphocytic leukemia. J. Clin. Oncol. 27, 4578–4584 apoptosis via Bak/Bax if Mcl-1 is neutralized. Cancer
107. Agathangelidis, A. et al. Highly similar genomic (2009). Cell 10, 389–399 (2006).
landscapes in monoclonal B cell lymphocytosis and 132. Abrisqueta, P. et al. Rituximab maintenance after 157. Blombery, P. et al. Acquisition of the recurrent
ultra-​stable chronic lymphocytic leukemia with low first-line therapy with rituximab, fludarabine, Gly101Val mutation in BCL2 confers resistance
frequency of driver mutations. Haematologica 103, cyclophosphamide, and mitoxantrone (R-​FCM) for to venetoclax in patients with progressive chronic
865–873 (2018). chronic lymphocytic leukemia. Blood 122, lymphocytic leukemia. Blood 132 (Suppl. 1), LBA-7
108. Ojha, J. et al. Monoclonal B cell lymphocytosis is 3951–3959 (2013). (2018).
characterized by mutations in CLL putative driver genes 133. Byrd, J. C. et al. Ibrutinib versus ofatumumab in 158. National Comprehensive Cancer Network. Chronic
and clonal heterogeneity many years before disease previously treated chronic lymphoid leukemia. N. Engl. lymphocytic leukemia / small lymphocytic lymphoma.
progression. Leukemia 28, 2395–2398 (2014). J. Med. 371, 213–223 (2014). NCCN https://www.nccn.org/professionals/physician_
109. Barrio, S. et al. Genomic characterization of high-​count 134. Burger, J. A. et al. Ibrutinib as initial therapy for gls/pdf/cll.pdf (2019).
MBL cases indicates that early detection of driver patients with chronic lymphocytic leukemia. N. Engl. J. 159. Goede, V. et al. Obinutuzumab plus chlorambucil in
mutations and subclonal expansion are predictors Med. 373, 2425–2437 (2015). patients with CLL and coexisting conditions. N. Engl.
of adverse clinical outcome. Leukemia 31, 170–176 135. Furman, R. R. et al. Idelalisib and rituximab in relapsed J. Med. 370, 1101–1110 (2014).
(2017). chronic lymphocytic leukemia. N. Engl. J. Med. 370, 160. Woyach, J. A. et al. Ibrutinib regimens versus
110. Ding, L. et al. Clonal evolution in relapsed acute 997–1007 (2014). chemoimmunotherapy in older patients with untreated
myeloid leukaemia revealed by whole-​genome 136. Jones, J. A. et al. Efficacy and safety of idelalisib in CLL. N. Engl. J. Med. 379, 2517–2528 (2018).
sequencing. Nature 481, 506–510 (2012). combination with ofatumumab for previously treated 161. Moreno, C. et al. Ibrutinib plus obinutuzumab versus
111. Gerlinger, M. et al. Intratumor heterogeneity chronic lymphocytic leukaemia: an open-​label, chlorambucil plus obinutuzumab in first-​line treatment
and branched evolution revealed by multiregion randomised phase 3 trial. Lancet Haematol. 4, of chronic lymphocytic leukaemia (iLLUMINATE):
sequencing. N. Engl. J. Med. 366, 883–892 (2012). e114–e126 (2017). a multicentre, randomised, open-​label, phase 3 trial.
112. Shanafelt, T. D. et al. Karyotype evolution on 137. Roberts, A. W. et al. Targeting BCL2 with venetoclax in Lancet Oncol. 20, 43–56 (2019).
fluorescent in situ hybridization analysis is associated relapsed chronic lymphocytic leukemia. N. Engl. J. Med. 162. Shanafelt, T. et al. A randomized phase III study of
with short survival in patients with chronic lymphocytic 374, 311–322 (2016). ibrutinib (PCI-32765)-based therapy vs. standard
leukemia and is related to CD49d expression. J. Clin. 138. Stilgenbauer, S. et al. Venetoclax in relapsed or fludarabine, cyclophosphamide, and rituximab (FCR)
Oncol. 26, e5–e6 (2008). refractory chronic lymphocytic leukaemia with 17p chemoimmunotherapy in untreated younger patients
113. Rossi, D. & Gaidano, G. Richter syndrome: pathogenesis deletion: a multicentre, open-​label, phase 2 study. with chronic lymphocytic leukemia (CLL): a trial of the
and management. Semin. Oncol. 43, 311–319 (2016). Lancet Oncol. 17, 768–778 (2016). ECOG-​ACRIN Cancer Research Group (E1912). Blood
114. Rossi, D., Spina, V. & Gaidano, G. Biology and treatment 139. Amin, N. A. et al. A quantitative analysis of subclonal 132 (Suppl. 1), LBA-4 (2018).
of Richter syndrome. Blood 131, 2761–2772 (2018). and clonal gene mutations before and after therapy in 163. Brown, J. R. How I treat CLL patients with ibrutinib.
115. Miller, C. R. et al. Near-​tetraploidy is associated with chronic lymphocytic leukemia. Clin. Cancer Res. 22, Blood 131, 379–386 (2018).
Richter transformation in chronic lymphocytic 4525–4535 (2016). 164. Burger, J. A. & O’Brien, S. Evolution of CLL treatment
leukemia patients receiving ibrutinib. Blood Adv. 1, 140. Pozzo, F. et al. NOTCH1 mutations associate with low — from chemoimmunotherapy to targeted and
1584–1588 (2017). CD20 level in chronic lymphocytic leukemia: evidence individualized therapy. Nat. Rev. Clin. Oncol. 15,
116. Weissmann, S. et al. Prognostic impact and landscape for a NOTCH1 mutation-​driven epigenetic dysregulation. 510–527 (2018).
of NOTCH1 mutations in chronic lymphocytic leukemia Leukemia 30, 182–189 (2016). 165. Badoux, X. C. et al. Fludarabine, cyclophosphamide,
(CLL): a study on 852 patients. Leukemia 27, 141. Messina, M. et al. Genetic lesions associated with and rituximab chemoimmunotherapy is highly effective
2393–2396 (2013). chronic lymphocytic leukemia chemo-​refractoriness. treatment for relapsed patients with CLL. Blood 117,
117. Villamor, N. et al. NOTCH1 mutations identify a Blood 123, 2378–2388 (2014). 3016–3024 (2011).
genetic subgroup of chronic lymphocytic leukemia 142. Dreger, P. et al. TP53, SF3B1, and NOTCH1 mutations 166. Fischer, K. et al. Bendamustine combined with
patients with high risk of transformation and poor and outcome of allotransplantation for chronic rituximab in patients with relapsed and/or refractory
outcome. Leukemia 27, 1100–1106 (2013). lymphocytic leukemia: six-​year follow-​up of the chronic lymphocytic leukemia: a multicenter phase II
118. Wu, Y. et al. Therapeutic antibody targeting of individual GCLLSG CLL3X trial. Blood 121, 3284–3288 (2013). trial of the German Chronic Lymphocytic Leukemia
Notch receptors. Nature 464, 1052–1058 (2010). 143. Hendriks, R. W., Yuvaraj, S. & Kil, L. P. Targeting Study Group. J. Clin. Oncol. 29, 3559–3566 (2011).
119. Maddocks, K. J. et al. Etiology of ibrutinib therapy Bruton’s tyrosine kinase in B cell malignancies. 167. Seymour, J. F. et al. Venetoclax–rituximab in relapsed
discontinuation and outcomes in patients with chronic Nat. Rev. Cancer 14, 219–232 (2014). or refractory chronic lymphocytic leukemia. N. Engl.
lymphocytic leukemia. JAMA Oncol. 1, 80–87 (2015). 144. Farooqui, M. Z. H. et al. Ibrutinib for previously J. Med. 378, 1107–1120 (2018).
120. Innocenti, I. et al. Clinical, pathological, and biological untreated and relapsed or refractory chronic 168. Jones, J. A. et al. Venetoclax for chronic lymphocytic
characterization of Richter syndrome developing after lymphocytic leukaemia with TP53 aberrations: leukaemia progressing after ibrutinib: an interim

Nature Reviews | Clinical Oncology


Reviews

analysis of a multicentre, open-​label, phase 2 trial. 178. Park, J. H. et al. A phase I first-​in-human clinical trial 190. Herling, C. D. et al. Complex karyotypes and KRAS
Lancet Oncol. 19, 65–75 (2018). of CD19-targeted 19-28z/4-1BBL ‘armored’ CAR and POT1 mutations impact outcome in CLL after
169. Byrd, J. C. et al. Acalabrutinib (ACP-196) in relapsed T cells in patients with relapsed or refractory NHL and chlorambucil-​based chemotherapy or
chronic lymphocytic leukemia. N. Engl. J. Med. 374, CLL including Richter’s transformation. Blood 132, chemoimmunotherapy. Blood 128, 395–404 (2016).
323–332 (2016). 224–224 (2018). 191. O’Brien, S. et al. Ibrutinib for patients with relapsed
170. Fabian, C. A. et al. SNS-062 demonstrates efficacy 179. Hallek, M. Chronic lymphocytic leukemia: or refractory chronic lymphocytic leukaemia with 17p
in chronic lymphocytic leukemia in vitro and inhibits 2017 update on diagnosis, risk stratification, and deletion (RESONATE-17): a phase 2, open-​label,
C481S mutated Bruton tyrosine kinase [abstract treatment. Am. J. Hematol. 92, 946–965 (2017). multicentre study. Lancet Oncol. 17, 1409–1418
1207]. Cancer Res. 77, 1207–1207 (2017). 180. Hillmen, P. et al. Initial results of ibrutinib plus (2016).
171. Jebaraj, B. M. C. et al. Vecabrutinib is efficacious venetoclax in relapsed, refractory CLL (Bloodwise TAP 192. Leonard, J. P. et al. Clinical activity of idelalisib
in vivo in a preclinical CLL adoptive transfer model. CLARITY Study): high rates of overall response, complete (GS-1101), a selective inhibitor of PI3Kδ, in phase 1
Blood 132, 1868–1868 (2018). remission and MRD eradication after 6 months of and 2 trials in chronic lymphocytic leukemia (CLL):
172. Allan, J. N. et al. Preliminary safety, pharmacokinetic, combination therapy. Blood 130, 428 (2017). effect of Del(17p)/TP53 mutation, Del(11q), IGHV
and pharmacodynamic results from a phase 1b/2 181. Fischer, K. et al. Venetoclax and obinutuzumab mutation, and NOTCH1 mutation. Blood 122,
dose-​escalation and cohort-​expansion study of the in chronic lymphocytic leukemia. Blood 129, 1632–1632 (2013).
noncovalent, reversible Bruton’s tyrosine kinase 2702–2705 (2017). 193. Oscier, D. G. et al. The clinical significance of NOTCH1
inhibitor (BTKi), vecabrutinib, in B-​lymphoid 182. Cramer, P. et al. Bendamustine followed by and SF3B1 mutations in the UK LRF CLL4 trial. Blood
malignancies. Blood 132, 3141–3141 (2018). obinutuzumab and venetoclax in chronic lymphocytic 121, 468–475 (2013).
173. de Weerdt, I., Koopmans, S. M., Kater, A. P. leukaemia (CLL2-BAG): primary endpoint analysis of a
& van Gelder, M. Incidence and management of multicentre, open-​label, phase 2 trial. Lancet Oncol. Acknowledgements
toxicity associated with ibrutinib and idelalisib: 19, 1215–1228 (2018). The work of the authors is supported by grants to F.B. from
a practical approach. Haematologica 102, 183. Jain, N. et al. Combined venetoclax and ibrutinib for the Ministry of Economy and Competitiveness, Instituto de
1629–1639 (2017). patients with previously untreated high-​risk CLL, and Salud Carlos III (17/00943) and to R.D.-F. from the US
174. Flinn, I. W. et al. The phase 3 DUO trial: duvelisib relapsed/refractory CLL: a phase II trial. Blood 130, Department of Health & Human Services, NIH, National
versus ofatumumab in relapsed and refractory CLL/SLL. 429–429 (2017). Cancer Institute (NCI) (1R35CA210105).
Blood 132, 2446–2455 (2018). 184. Byrd, J. C. et al. Targeting BTK with ibrutinib in
175. Younes, A. et al. Safety and activity of ibrutinib in relapsed chronic lymphocytic leukemia. N. Engl. Author contributions
combination with nivolumab in patients with relapsed J. Med. 369, 32–42 (2013). The authors contributed equally to all aspects of the article.
non-​Hodgkin lymphoma or chronic lymphocytic 185. Mansouri, L. et al. NOTCH1 and SF3B1 mutations can
leukaemia: a phase 1/2a study. Lancet Haematol. 6,
Competing interests
be added to the hierarchical prognostic classification
The authors declare no competing interests.
e67–e78 (2019). in chronic lymphocytic leukemia. Leukemia 27,
176. Mato, A. R. et al. Phase I/II study of umbralisib (TGR- 512–514 (2013). Publisher’s note
1202) in combination with ublituximab (TG-1101) and 186. Wierda, W. G. et al. Multivariable model for time to Springer Nature remains neutral with regard to jurisdictional
pembrolizumab in patients with relapsed/refractory first treatment in patients with chronic lymphocytic claims in published maps and institutional affiliations.
CLL and Richter’s transformation. Blood 132, 297 leukemia. J. Clin. Oncol. 29, 4088–4095 (2011).
(2018). 187. Rossi, D. et al. Alteration of BIRC3 and multiple Reviewer information
177. Cwynarski, K. et al. Autologous and allogeneic stem-​cell other NF-​κB pathway genes in splenic marginal zone Nature Reviews Clinical Oncology thanks S. Stilgenbauer,
transplantation for transformed chronic lymphocytic lymphoma. Blood 118, 4930–4934 (2011). J. Burger and M. Hallek, for their contribution to the peer
leukemia (Richter’s syndrome): a retrospective analysis 188. Baliakas, P. et al. Prognostic relevance of MYD88 review of this work.
from the chronic lymphocytic leukemia subcommittee mutations in CLL: the jury is still out. Blood 126,
of the chronic leukemia working party and lymphoma 1043–1044 (2015).
working party of the European Group for Blood and 189. Hillmen, P. et al. Alemtuzumab compared with Related links
Marrow Transplantation. J. Clin. Oncol. 30, chlorambucil as first-​line therapy for chronic lymphocytic ClinicalTrials.gov: https://clinicaltrials.gov/
2211–2217 (2012). leukemia. J. Clin. Oncol. 25, 5616–5623 (2007).

www.nature.com/nrclinonc

You might also like