ArticlePDF AvailableLiterature Review

Aggressive NK Cell Leukemia: Current State of the Art

MDPI
Cancers
Authors:

Abstract and Figures

Simple Summary Aggressive natural killer cell leukemia (ANKL) is a rare, lethal disease that presents many diagnostic and therapeutic challenges. Recent studies have shed new light on the salient features of its molecular pathogenesis and provided further insight into the clinicopathologic spectrum of this disease. This review presents a state-of-the-art overview of ANKL, spanning its historical evolution as a distinct entity, pathobiology, and potential therapeutic vulnerabilities. Abstract Aggressive natural killer (NK) cell leukemia (ANKL) is a rare disease with a grave prognosis. Patients commonly present acutely with fever, constitutional symptoms, hepatosplenomegaly, and often disseminated intravascular coagulation or hemophagocytic syndrome. This acute clinical presentation and the variable pathologic and immunophenotypic features of ANKL overlap with other diagnostic entities, making it challenging to establish a timely and accurate diagnosis of ANKL. Since its original recognition in 1986, substantial progress in understanding this disease using traditional pathologic approaches has improved diagnostic accuracy. This progress, in turn, has facilitated the performance of recent high-throughput studies that have yielded insights into pathogenesis. Molecular abnormalities that occur in ANKL can be divided into three major groups: JAK/STAT pathway activation, epigenetic dysregulation, and impairment of TP53 and DNA repair. These high-throughput data also have provided potential therapeutic targets that promise to improve therapy and outcomes for patients with ANKL. In this review, we provide a historical context of the conception and evolution of ANKL as a disease entity, we highlight advances in diagnostic criteria to recognize this disease, and we review recent understanding of pathogenesis as well as biomarker discoveries that are providing groundwork for innovative therapies.
Content may be subject to copyright.
cancers
Review
Aggressive NK Cell Leukemia: Current State of
the Art
Siba El Hussein , L. Jerey Medeiros and Joseph D. Khoury *
Department of Hematopathology, the University of Texas MD Anderson Cancer Center,
Houston, TX 77030, USA; sel2@mdanderson.org (S.E.H.); ljmedeiros@mdanderson.org (L.J.M.)
*Correspondence: Jkhoury@mdanderson.org
Received: 26 August 2020; Accepted: 29 September 2020; Published: 9 October 2020


Simple Summary:
Aggressive natural killer cell leukemia (ANKL) is a rare, lethal disease that
presents many diagnostic and therapeutic challenges. Recent studies have shed new light on the
salient features of its molecular pathogenesis and provided further insight into the clinicopathologic
spectrum of this disease. This review presents a state-of-the-art overview of ANKL, spanning its
historical evolution as a distinct entity, pathobiology, and potential therapeutic vulnerabilities.
Abstract:
Aggressive natural killer (NK) cell leukemia (ANKL) is a rare disease with a grave prognosis.
Patients commonly present acutely with fever, constitutional symptoms, hepatosplenomegaly,
and often disseminated intravascular coagulation or hemophagocytic syndrome. This acute clinical
presentation and the variable pathologic and immunophenotypic features of ANKL overlap with
other diagnostic entities, making it challenging to establish a timely and accurate diagnosis of
ANKL. Since its original recognition in 1986, substantial progress in understanding this disease
using traditional pathologic approaches has improved diagnostic accuracy. This progress, in turn,
has facilitated the performance of recent high-throughput studies that have yielded insights into
pathogenesis. Molecular abnormalities that occur in ANKL can be divided into three major groups:
JAK/STAT pathway activation, epigenetic dysregulation, and impairment of TP53 and DNA repair.
These high-throughput data also have provided potential therapeutic targets that promise to improve
therapy and outcomes for patients with ANKL. In this review, we provide a historical context of the
conception and evolution of ANKL as a disease entity, we highlight advances in diagnostic criteria to
recognize this disease, and we review recent understanding of pathogenesis as well as biomarker
discoveries that are providing groundwork for innovative therapies.
Keywords: leukemia; NK cell; molecular; immunohistochemistry; biomarker
1. Introduction
Aggressive natural killer (NK) cell leukemia (ANKL) is a rare, fulminant disease with a dismal
prognosis that presents many diagnostic challenges. Since its original description in the 1980s,
the diagnostic approach to ANKL has evolved slowly, in large part due to disease rarity and the
complexity of clinical and pathologic presentations. An additional major challenge has been a lack
of a specific immunophenotypic or molecular signature characteristic of ANKL. Most early studies
attempted to provide a systematic approach to the diagnosis ANKL by relying on clinical and
morphologic correlation, coupled with ancillary tools such as flow cytometry and conventional
karyotyping. Although robust, these tools did not uncover specific biomarkers, partly because
traditional approaches lack the versatility inherent in next-generation sequencing, particularly in the
context of an NK cell neoplasm.
Cancers 2020,12, 2900; doi:10.3390/cancers12102900 www.mdpi.com/journal/cancers
Cancers 2020,12, 2900 2 of 15
In recent years, high-throughput genomic analyses at the DNA and RNA levels has contributed
greatly to our understanding of ANKL and provided novel grounds for potential targeted therapies.
This review is intended to provide an overview of ANKL, as defined in the current World
Health Organization (WHO) classification [
1
], spanning its historic origin, clinical and laboratory
manifestations, biology, and novel therapeutic approaches, with emphasis on advances in our
understanding of this rare malignancy in light of recent advances. This review is based on a review of
the literature using the term “aggressive NK cell leukemia”.
2. Historical Overview
The recognition of ANKL as a separate entity was presaged by the disease group referred to
as NK-type lymphoproliferative disease of granular lymphocytes (NK-LDGL). Entailing abnormal
proliferation of large granular lymphocytes (LGLs) expressing a characteristic NK cell immunophenotype
(CD3
/CD56+/CD16+), NK-LGDL was understood to encompass heterogeneous clinical manifestations
ranging from indolent, asymptomatic conditions to aggressive, fulminant diseases. As a body of
informative work underscoring some characteristics of NK-LGDL emerged [
2
5
], gradual attention was
turned to separating indolent forms from those with an acute clinical course. Thus eventually emerged
from NK-LDGL the entities of NK-LGL, chronic lymphoproliferative disorder of NK cells (CLPD-NK),
and ANKL.
The term ANKL was first mentioned in a publication by Fernandez et al. in 1986 [
6
]. The authors
described a case of a 70-year-old man with multiple episodes of intestinal perforation of unknown
cause, splenomegaly, and bone marrow infiltration by cells described as being morphologically similar
to large granular lymphocytes (LGLs), but lacking T cell markers. The patient had a rapidly progressive
clinical course and died within two months of hospital admission. At autopsy, he had extensive
involvement of the bone marrow, spleen, lymph nodes, and jejunum by LGL-like cells. Conventional
cytogenetic analysis revealed clonal aberrations, leading the authors to conclude that the disease
was neoplastic. The authors also postulated that this disease represented an aggressive form of NK
cell leukemia.
In 1988, Imamura et al. reported two cases of NK cell leukemia/lymphoma that lacked clonal
rearrangements of the T-cell receptor (TRB,TRG) and immunoglobulin genes [
7
]. The authors noted that
the neoplastic cells in these two cases expressed CD56 (NKH-1/Leu-19) and lacked CD57 (HNK-1/Leu-7).
Two years later, the same authors compared four cases of ANKL to seven other similar cases reported
in the literature [
6
,
8
11
]. Imamura and colleagues concluded that this disease emanates from “a third
lineage of lymphoid cells” and used the term “aggressive natural killer cell leukemia/lymphoma” [
12
].
ANKL was subsequently reported to arise in two patients with “lethal midline granuloma”,
currently known as extra-nodal NK/T cell lymphoma, nasal type (ENKTL), raising the possibility
that ANKL disease could represent a terminal event in the evolution of ENKTL [
13
]. This report was
followed by another, which described four cases of ANKL with dissemination to various organs in a
manner akin to lymphomas [
14
]. These reports raised controversy regarding the relationship between
ANKL and ENKTL, which arguably remains largely unresolved [15,16].
In 1994, the Revised European–American classification of lymphoid neoplasms recognized ANKL
as an entity separate from ENKTL, and such separation was adopted in the WHO classification
system [17,18].
3. Clinical Features
ANKL patients have a median age of 40 years, and there is no gender predilection. The disease has a
higher incidence in Asian populations, but ANKL can also arise in other groups [
19
]. The most common
presenting signs and symptoms include fever, B-symptoms, hepatosplenomegaly, lymphadenopathy,
disseminated intravascular coagulopathy, and hemophagocytosis. Epstein-Barr virus (EBV) infection
has been observed in a subset of ANKL cases. Despite intensive chemotherapy, patients with ANKL
have a very poor prognosis, with a median survival of less than two months [
20
,
21
]. Several prognostic
Cancers 2020,12, 2900 3 of 15
factors have been proposed, including patient age, serum lactate dehydrogenase level, and serum total
bilirubin level [
22
], but these factors have not been validated [
23
]. The clinicopathologic features of
ANKL have been elucidated in several case series [1921,2427].
4. Morphologic Features
Histologically, bone marrow involvement by ANKL can be prominent or subtle, with two main
patterns of infiltration: interstitial and sinusoidal [
19
]. The neoplastic cells are generally medium-sized,
possess a moderate amount of cytoplasm, and exhibit highly irregular nuclei with condensed chromatin
and conspicuous nucleoli. Apoptosis and focal necrosis are common findings; however, geographic
necrosis is typically not observed in ANKL, unlike ENKTL [
19
] (Figure 1A). Microscopic examination
of peripheral blood and bone marrow smear preparations usually demonstrate neoplastic cells that
are intermediate to large in size, with moderate amounts of basophilic agranular cytoplasm often
containing punched-out vacuoles. The neoplastic cells typically have highly irregular nuclear contours
and an open chromatin pattern with prominent nucleoli.
Cancers 2020, 12, x FOR PEER REVIEW 3 of 16
dehydrogenase level, and serum total bilirubin level [22], but these factors have not been validated
[23]. The clinicopathologic features of ANKL have been elucidated in several case series [19–21,24
27].
4. Morphologic Features
Histologically, bone marrow involvement by ANKL can be prominent or subtle, with two main
patterns of infiltration: interstitial and sinusoidal [19]. The neoplastic cells are generally medium-
sized, possess a moderate amount of cytoplasm, and exhibit highly irregular nuclei with condensed
chromatin and conspicuous nucleoli. Apoptosis and focal necrosis are common findings; however,
geographic necrosis is typically not observed in ANKL, unlike ENKTL [19] (Figure 1A). Microscopic
examination of peripheral blood and bone marrow smear preparations usually demonstrate
neoplastic cells that are intermediate to large in size, with moderate amounts of basophilic agranular
cytoplasm often containing punched-out vacuoles. The neoplastic cells typically have highly
irregular nuclear contours and an open chromatin pattern with prominent nucleoli.
Figure 1. Aggressive natural killer (NK) cell leukemia involving bone marrow. (A) Trephine biopsy
showing an extensive infiltrate of neoplastic lymphocytes in interstitial and less pronounced
sinusoidal infiltration patterns. (B) CD2 immunohistochemistry, positive in neoplastic cells. (C)
Perforin A immunohistochemistry, positive in neoplastic cells. (D) Epstein–Barr virus encoded small
RNA (EBER1/2) expression in neoplastic cells. (E) Flow cytometry immunophenotyping performed
on an aspirate sample demonstrated an aberrant population of lymphocytes (pink highlight), positive
for CD2, CD8, CD16, and CD56. These cells were negative for CD3 and CD5. In addition, they were
negative for CD4, CD57, T cell receptor (TCR)-alpha/beta, and TCR-gamma/delta.
5. Immunophenotypic Features
When assessing normal NK cells by multiparameter/multicolor flow cytometry
immunophenotyping, 90% were CD56dim/CD16bright, and 10% were CD56bright/CD16dim [27]. It is
thought that the CD56dim/CD16bright group represents a more mature stage of NK cell differentiation,
but prior to mature NK cells with expression of CD57 and killer immunoglobulin-like receptors
(KIRs). The CD56bright/CD16dim group represents an earlier stage of differentiation [19,28,29].
Following this rationale, NK cell neoplasms can be divided into two broad groups. The first group is
CD56bright/posit ive/CD16dim/negative, suggesting failure of differentiation [30]. This group, which
encompasses ANKL and ENKTL, retains a cytokine-secreting function that underlies the cytokine
storm that occurs characteristically in patients with these two diseases [30]. The second group is
Figure 1.
Aggressive natural killer (NK) cell leukemia involving bone marrow. (
A
) Trephine biopsy
showing an extensive infiltrate of neoplastic lymphocytes in interstitial and less pronounced sinusoidal
infiltration patterns. (
B
) CD2 immunohistochemistry, positive in neoplastic cells. (
C
) Perforin A
immunohistochemistry, positive in neoplastic cells. (
D
) Epstein–Barr virus encoded small RNA
(EBER1/2) expression in neoplastic cells. (
E
) Flow cytometry immunophenotyping performed on an
aspirate sample demonstrated an aberrant population of lymphocytes (pink highlight), positive for
CD2, CD8, CD16, and CD56. These cells were negative for CD3 and CD5. In addition, they were
negative for CD4, CD57, T cell receptor (TCR)-alpha/beta, and TCR-gamma/delta.
5. Immunophenotypic Features
When assessing normal NK cells by multiparameter/multicolor flow cytometry immunophenotyping,
90% were CD56
dim
/CD16
bright
, and 10% were CD56
bright
/CD16
dim
[
27
]. It is thought that
the CD56
dim
/CD16
bright
group represents a more mature stage of NK cell dierentiation,
but prior to mature NK cells with expression of CD57 and killer immunoglobulin-like receptors
(KIRs). The CD56
bright
/CD16
dim
group represents an earlier stage of dierentiation [
19
,
28
,
29
].
Following this rationale, NK cell neoplasms can be divided into two broad groups. The first
group is CD56
bright/positive
/CD16
dim/negative
, suggesting failure of dierentiation [
30
]. This group,
which encompasses ANKL and ENKTL, retains a cytokine-secreting function that underlies the
cytokine storm that occurs characteristically in patients with these two diseases [
30
]. The second group
Cancers 2020,12, 2900 4 of 15
is CD56
dim/negative
/CD16
bright/positive
, encompassing chronic lymphoproliferative disorder of NK cells
(CLPD-NK), and characterized by neoplastic cells with a cytotoxic function [30,31] (Table 1).
Table 1.
An integrative clinicopathologic overview, comparing multiple aspects of NK cell proliferative disorders.
Diagnosis Etiology Immunophenotype Functionality Genomic Landscape
Transient increase
in NK cells
Autoimmune disorders,
viral infections
CLPD* Unknown stimulus,
possibly viral
CD8+(uniform
positivity), CD16+,
CD56+(dim); Loss of
CD2, CD7 and CD57
Cytotoxic function Activating mutations in the
STAT3 SH2 domain
ENKTL* Strong association
with EBV infection
CD2+, CD56+; Loss of
sCD3, CD4, CD8, CD16
and CD57
Cytokine secretion
function
DDX3X, JAK/STAT signaling
pathway (STAT3, STAT5B,
JAK3, and PTPRK), KIT,
CTNNB1, TP53, PRDM1,
ATG5, AIM1,FOX03,
and HACE1, RAS, MYC,
KMT2D/MLL2, ARID1A,
EP300, ASXL3, CDKN2A,
CDKN2B, CDKN1A, FAS
ANKL* Strong association
with EBV infection
CD2+, CD16+, CD56+;
Loss of sCD3, CD4, CD7,
CD8 and CD57
Cytokine secretion
function
JAK/STAT signaling pathway,
TP53, TET2, CREBBP, ASXL1,
ASXL2, BRINP3, PRPF40B
*CLPD: chronic lymphoproliferative disorder; *ENKTL: extra-nodal NK/T cell lymphoma; *ANKL: aggressive NK
cell leukemia.
The main flow cytometry features of ANKL are prominent forward scatter (FSC), denoting
an increased cell size when compared to non-neoplastic background lymphocytes, and expression
of CD56 and CD94 [
19
]. CD56 is a neural cell adhesion molecule that assists in infiltration and
invasion. CD94 is a molecule that forms heterodimers with either NKG2A (CD159a) or NKG2C
(CD159c). The lectin-like CD94/NKG2A receptor is inhibitory and recognizes the human leukocyte
antigen (HLA) class Ib molecule HLA-E as its predominant ligand [
32
,
33
], while the CD94/NKG2C is
activating/triggering [
34
,
35
]. While Zambello et al. have demonstrated that in the vast majority of
NK-LGDL cases (16/18; 88.9%) CD94 is associated with NKG2A [
5
], to our knowledge the functional
associations of CD94 in ANKL has not been characterized yet. ANKL cells appear to consistently
express CD2, in addition to cytoplasmic CD3 (epsilon chain), CD16, and cytotoxic molecules, such as
granzyme B, TIA1, and perforin A. Expression of CD7 or CD8 is heterogeneous, whereas CD3 (surface),
CD4, CD5, CD57, and T cell receptors (TCR) are usually absent in ANKL [
36
]. The KIR (killer cell
immunoglobulin-like receptor) proteins (CD158a/b/e), known to be present only in peripheral blood
CD56
dim
NK cells, are mostly negative in ANKL [
19
,
36
]. In practical terms, neoplastic NK cell
populations can be separated from lymphocytes using a CD45/SSC gating strategy. In the case of
neoplastic NK cells overlapping with lymphocytes, using a CD56/CD3 gating strategy is helpful to
identify the aberrant cells [36] (Figure 1B–E and Table 2).
Table 2. Immunophenotypic characteristics of aggressive NK cell leukemia.
Consistently Positive Markers Frequently Positive Markers Frequently Negative Markers
CD2 CD7 CD3
Cytoplasmic CD3 epsilon CD8 CD4
CD16 CD5
CD56 CD57
CD94 KIR (CD158a–e)
Granzyme B T-cell receptors
TIA
Perforin
One challenge to establishing an early diagnosis of ANKL is the common presence of only a
small number of neoplastic NK cells in the bone marrow [
19
]. In one study of ANKL, the proportion
Cancers 2020,12, 2900 5 of 15
of abnormal NK cells in the bone marrow was reported to be <5% in half of the patients [
27
].
Flow cytometry immunophenotypic analysis is particularly useful in the early detection of ANKL,
usually with a high specificity [
27
]. As ANKL progresses, the number of neoplastic cells in the bone
marrow increases. In one study, the rate of detection of aberrant NK cell populations increased
from 86.8% to 97.4% when second and third bone marrow samples were assessed as the disease
progressed [
27
]. Thus, it is advisable to obtain several specimens from the blood, bone marrow, or other
tissue sites and to conduct consecutive flow cytometry analyses when positivity is not detected and
the patient is suspected clinically to have a neoplasm such as ANKL [27] (Figure 1E).
Homing of neoplastic NK cells to body cavities has been outlined recently in NK cell
neoplasms, including ANKL, creating a dierential diagnosis with primary eusion lymphoma [
37
40
].
This dierential diagnosis can be addressed successfully by using flow cytometry immunophenotyping
when morphologic examination is impeded or is of limited value [36].
6. The Role of Epstein–Barr Virus
The first cases of ANKL reported by Fernandez et al. and Imamura et al. were EBV-negative or
EBV was not assessed [
6
,
12
]. Kawa-Ha et al., in 1989, were the first to highlight that neoplastic NK cells
in the “aggressive” and “chronic” forms of “lymphoproliferative disease of granular lymphocytes”
(currently, ANKL and T cell LGL, respectively) could carry EBV DNA, thus implicating EBV in the
pathogenesis of these diseases [
41
]. EBV-positive cases are positive for EBV-encoded RNA (EBER) and
are negative for latent membrane protein type 1 (LMP-1) in some reports, suggesting a type I latency
pattern of infection [
24
,
25
]. This latency pattern is believed to provide an advantage to neoplastic NK
cells by enabling them to evade host virus-specific cytotoxic T cell activity [42,43].
Although ANKL has an established association with EBV infection, EBV-negative ANKL is also
widely recognized [
19
21
,
44
48
]. In contrast with EBV-positive ANKL, EBV-negative cases occur
more frequently in older patients and arise equally in Asian and non-Asian
populations [44,45]
.
No morphologic or immunophenotypic dierences have been shown in EBV-positive versus
EBV-negative ANKL [
45
]. Although some reports have suggested a more indolent behavior of
EBV-negative ANKL [
46
,
49
,
50
], recent articles have shown that these cases are associated with an
aggressive clinical behavior, similar to EBV-positive cases [44,45].
The absence of EBV in a subset of ANKL cases can potentially lead to a delayed diagnosis,
with adverse clinical consequences. When evaluating a potential case of ANKL, one should not use
EBV-negative status as a criterion to exclude the diagnosis. Further investigation of these neoplasms is
needed to unmask NK cell lineage. Performing immunohistochemical (IHC) analysis can be helpful
because CD56 and cytotoxic molecules are reported to be positive in most ANKL cases [44,45].
7. Cytogenetic Features
Conventional cytogenetic abnormalities in ANKL have been reported in few studies, and these
abnormalities commonly include del (6) (q21q25) and del (11q) [
24
,
25
,
51
]. Another study found an
association between ANKL and chromosome 7 abnormalities, and also reported chromosome 6q
deletion associated with ENKTL [
52
]. More recently, array-based comparative genomic hybridization
(aCGH) analysis in a cohort of patients with ANKL and ENKTL showed that gains of 1q23.1–q23.2 and
1q31.3–q44, as well as losses of 7p15.1–q22.3 and 17p13.1, are more frequent in ANKL than ENKTL [
53
].
In contrast, cases of ENKTL more frequently had gains of 2q and losses of 6q16.1–q27, 11q22.3–q23.3,
5p14.1–p14.3, 5q34–q35.3, 1p36.23–p36.33, 2p16.1–p16.3, 4q12, and 4q31.3–q32.1 [
53
]. Others have
reported that loss of 6q16.1–q27 is a common finding in NK cell malignancies [
54
], detected in both
ANKL [24,25,51] and ENKTL, but more frequently in ENKTL [53].
8. Molecular Pathogenesis and Genomic Landscape
Abundant data are available with regards to genetic pathways involved in the pathogenesis of
ENKTL. These abnormalities include mutations in the JAK/STAT [
55
59
], AKT [
59
], and NF-
κ
B [
59
]
Cancers 2020,12, 2900 6 of 15
signaling pathways. Additional abnormalities include recurrent chromosomal aberrations in ENKTL
including a 6q21 deletion [
60
] (silencing the tumor suppressors PRDM1 and FOXO310), and mutations
in the RNA helicase gene DDX3X [
61
]. However, apart from copy number aberration analyses [
24
,
25
,
53
],
a deeper understanding of the genetic alterations in ANKL was missing until recently. The advent of
next generation sequencing methods has provided an unprecedented impetus for understanding the
molecular pathogenesis of ANKL. Three recent studies [
19
,
62
,
63
] provided a comprehensive genetic
analysis of ANKL via next generation sequencing (Table 3). While the findings in these studies show
some dierences that are likely the result of diering methodologies, they share many common threads
that provide insight into the molecular landscape of ANKL. Major genetic findings in these studies are
discussed below:
Table 3.
Breakdown of the common mutations reported in the three most recent genomic studies of
aggressive NK cell leukemia.
Mutations Huang et al. [63]
(29 Patients)
Dufva et al. [62]
(14 Patients)
El Hussein et al. [19]
(6 Patients)
JAK/STAT
STAT3, STAT5B, STAT5A,
JAK2, JAK3, STAT6,
SOCS1, SOCS3 and
PTPN11 (48%)
STAT3 (21%) JAK1, JAK3, STAT3
(66.6%)
RAS/MAPK (29%) (16.7%)
Epigenetic modifiers
TET2 (28%), CREBBP
(21%), KMT2D (21%),
BCOR (3%)
SETD2, KMT2D and
BCOR (50%), TET2 (7%)
TET2 (16.7%), CREBBP
(16.7%), GFI1 (16.7%)
RNA helicase (DDX3X) (7%) (21%)
Cell cycle regulation and
DNA damage repair TP53 (34%) TP53 (7%)
TP53 (50%), ASXL1
(33.3%), ASXL2 (33.3%),
BRINP3 (16.7%)
mRNA splicing PRPF40B (16.7%)
8.1. JAK/STAT Signaling Pathway
Four JAKs (JAK1, JAK2, JAK3, TYK2) and seven STATs (STAT1, STAT2, STAT3, STAT4, STAT5a,
STAT5b, STAT6) are used by more than 50 cytokines and growth factors [
64
]. Extracellular binding of
cytokines or growth factors with their corresponding trans-membrane receptors induce conformational
changes in receptor-bound JAK proteins, creating a distance between their kinase domains and
inhibitory pseudo-kinase domains [
65
]. Trans-activated JAKproteins subsequently phosphorylate STAT
proteins, resulting in dimerization, nuclear translocation, and direct DNA binding [
65
]. STATs disperse
throughout the genome and regulate transcription of both protein-coding and non-coding genes [
66
].
In addition, all STATs recognize the same DNA sequence, known as the GAS motif [
64
]. However,
STATs may antagonize the action of one another by competing for the binding of the same genomic
site [
64
]. For example, in T cells, dendritic cells, and cancer cell lines, STAT3-driven IL-17 transcription
is blocked by STAT5 [6769].
Dufva et al. analyzed 14 patients of ANKL using whole-exome sequencing [
62
]. They showed
frequent genetic mutations in the JAK/STAT (21% of cases exhibited STAT3 mutations) and RAS-MAPK
signaling pathways [
62
]. Similarly, Huang et al. analyzed eight patients with ANKL by whole-genome
sequencing (WGS) and 29 ANKL (including the eight patients analyzed by WGS) patients by targeted
sequencing [
63
]. They noted that mutations in molecules of the JAK/STAT system, namely, STAT3,
STAT5B,STAT5A,JAK2,JAK3,STAT6,SOCS1,SOCS3, and PTPN11, were seen in 48% of patients,
with 17% of the cases harboring STAT3 mutations [
63
]. El Hussein et al. analyzed six ANKL patients,
finding mutations in JAK1 in one patient (who also harbored a mutation in STAT3), JAK3 in one
patient, and STAT3 in three patients [
19
]. In general, mutations in the JAK/STAT pathway were
mutually exclusive.
Cancers 2020,12, 2900 7 of 15
Huang et al. [
63
] and Dufva et al. [
62
] found that most of the STAT3 and STAT5B mutations
were localized to exons 20 and 21 encoding the Src homology 2 (SH2) domain, which mediates STAT
protein dimerization [
63
]. This domain also constitutes the hotspot containing activating mutations in
NKTCL [
55
,
61
]. Other JAK/STAT-related mutations reported were 9p copy number gain-containing
JAK2 and a point mutation in the protein tyrosine phosphatase (PTP) PTPRK, as well as mutations
in PTPN4 and PTPN23 [
62
]. Of these molecules, PTPRK is a tumor suppressor shown to negatively
regulate STAT3, and is commonly deleted in NKTCL.
Huang et al. suggested that EBV-encoded small RNAs (EBERs) that are highly expressed
in some cases of ANKL induce the release of massive amounts of IL-10, a well-known upstream
activator of the JAK/STAT pathway [
70
]. This stimulation in turn stimulates STAT3 phosphorylation,
leading to downstream MYC activation [
63
]. Importantly, Huang and colleagues also observed
that JAK/STAT-mutated and -unmutated leukemic NK cells showed a similar expression pattern
in MYC-driven programs [
63
]. This finding suggests that STAT3 signaling can be activated
by over-production of IL-10 or through other unknown mechanisms, independent of genetic
mutations [
63
]. This observation has been supported by immunohistochemical analysis, which has
shown phosphorylated STAT3 (p-STAT3; indicative of activation) is significantly higher in neoplastic
cells residing in the bone marrow of both JAK/STAT wild-type and mutated ANKL cases as compared
with controls [63].
The high frequency of mutations involving the JAK/STAT signaling pathway prompted Huang et al.
to analyze the plasma levels of inflammatory cytokines in patients with ANKL [
63
]. They found
significantly high serum levels of IL-10 in ANKL patients [
63
]. The authors concluded that IL-10 plays
a major role in upstream activation of the JAK/STAT pathway in ANKL, leading to increased expression
of MYC [
63
]. The authors subsequently proposed an IL-10–STAT3–MYC transcription regulation model
involved in the pathogenesis of ANKL [
63
]. On another note, IL-10 treatment of cell lines in the same
study preferentially stimulated JAK/STAT wild-type cells, but not the STAT3 Y640F-mutant ANKL
cells, causing an increase pathway activity and cellular proliferation [
63
]. This result suggests that
IL-10 upregulation plays a more prominent role in JAK/STAT unmutated ANKL patients [63].
8.2. Epigenetic Dysregulation
Half of the cases in the Dufva et al. study harbored mutations in epigenetic regulatory molecules
such as TET2 and CREBBP, and four (28%) patients had mutations in the RNA helicase DDX3X [
62
].
Huang and colleagues also identified mutations in epigenetic modification-related genes, including
TET2 (28%), CREBBP (21%), and MLL2 (21%) [
63
]. In contrast, DDX3X and BOCR, commonly found in
ENKTL [
61
,
71
], were less frequently mutated in ANKL [
63
]. Our group also has found mutations in
TET2,CREBBP, and GFI1 in ANKL cases [19].
8.3. TP53 Alterations and DNA Repair
Huang et al. identified TP53 mutations in 34% of cases of ANKL [
63
], whereas Dufva and
colleagues identified TP53 mutations in 1 out of 14 patients [
62
]. In our study, we found TP53 mutations
in three of six patients, and demonstrated uniform absence of p53 protein expression in four of eight
ANKL cases assessed by immunohistochemistry [
19
]. This finding is in keeping with observations in
an earlier aCGH study by Nakashima et al., who reported 17p13.1 losses more frequently in ANKL
than ENKTL cases [53].
Dufva and colleagues noted the lack of a DNA double-strand break repair-associated signature
in ANKL cases when compared with ENKTL, CLPD-NK, and T-LGL, suggesting a divergence in
underlying mutational processes between ANKL and ENKTL [
62
]. However, no mutations specific
to ANKL were uncovered in any of the recent studies [
62
,
63
]. Moreover, several genes that have
been identified in ENKTL cases, such as DDX3X [
61
], STAT3 [
55
,
61
], BCOR [
71
], and KMT2D [
61
],
were also identified in ANKL [
62
,
63
]. From these observations, we suggest that one can infer that the
pathogenesis of ANKL and NKTCL is closely related. Nevertheless, mutations in genes such as DDX3X
Cancers 2020,12, 2900 8 of 15
and TP53 were reported to contribute to a poorer prognosis in ENKTL [
61
], whereas the distribution of
these same mutations was not homogeneous in recent studies of ANKL (Table 1). This observation
suggests that mutations DDX3X and TP53 play a minor role in ANKL and refutes the hypothesis
that ANKL evolves from ENKTL. It is also worth mentioning that although a connection between
EBV status and mutational signature was suggested in past studies [
44
], this theory appears to have
weakened because the investigation of additional ANKL cases have been frequently EBV-negative.
Several gene mutations identified in EBV-positive cases also have been identified in EBV-negative
cases, such as STAT3 [
45
], TP53 [
44
], TET2 [
44
], and DDX3X [
44
]. Therefore, although EBV plays an
undeniable role in the pathogenesis of a subset of cases of ANKL, other epigenetic factors may exert
similar alterations on the genomic level, resulting in ANKL independent of EBV.
9. Examples of Clinicogenomic Data Integration
An example of genomic and clinical data integration is illustrated by the work of Tang et al. [
22
].
These authors identified 29 cases of classic ANKL with a fulminant presentation and eight cases of ANKL
with “subacute clinical course”. The latter group was defined as patients who manifested infectious
mononucleosis-like symptoms for more than 90 days, before full-blown manifestations of ANKL
developed. This subacute group is important to recognize because it can be mistaken it for a self-limited
infectious disease, likely delaying intervention with adverse clinical consequences [
22
]. By applying
ultra-high multiplex PCR technology, they identified mutations in TP53 (most commonly), genes in the
JAK/STAT RAS-MAPK signal transduction systems, in the transcription factors NF-
κ
B1 and NF-
κ
B1A,
and in the epigenetic regulatory molecules TET2 and CREBBP [
22
]. TP53 was mutated significantly
less often in “subacute” ANKL (none were detected) in comparison to fulminant ANKL [
22
]. However,
gene mutations in the JAK/STAT pathway were similar between the two groups, suggesting that the
key driving mechanisms are similar between the classic and subacute variants of ANKL [22].
El Hussein et al. published an integrative genomic and immunophenotypic landscape study of
12 cases of ANKL, of which six cases had available next generation sequencing data [
19
]. The authors
also identified mutations in the JAK/STAT (JAK1,JAK3) and RAS/MAPK pathways (KRAS), as well as in
epigenetic modifiers (TET2,CREBBP, and GFI1), cell cycle regulation, and DNA damage repair (TP53,
ASXL1,ASXL2, and BRINP3) and mRNA splicing factors (PRPF40B) (Table 3). In addition, the authors
coupled these genomic findings with immunophenotypic analysis by immunohistochemistry, showing
frequent loss of p53 expression, in addition to overexpression of BCL-2 and MYC in ANKL cases. To date,
this study includes the largest number of cases at a single institution analyzed by next-generation
sequencing performed from a clinical perspective, as other studies, although highly valuable,
were performed more from translational and investigational perspectives.
10. Immune Checkpoint Status
Gao et al. reported CD274 (PD-L1) overexpression in two of three ANKL cases [
44
]. Furthermore,
our group has shown PD-L1 expression in two of eight ANKL cases [
19
]. In a small group of patients with
ANKL and ENKTL, JAK2 gain was associated with gains of the neighboring immune evasion-associated
CD274 (PD-L1) and CD273 (PD-L2) genes [
62
]. In addition, sensitivity of ENKTL to PD-1 inhibition
was recently outlined [
72
]. Taking all of these data into account, PD-1 inhibition represents a promising
approach for ANKL treatment. This may be particularly true in EBV-positive cases of ANKL, as EBV
infection plays a role in PD-L1 stimulation [73].
11. Treatment of ANKL
11.1. Chemotherapy in ANKL
No consensus chemotherapeutic regimen has been established to manage patients with ANKL as
the rarity of this disease has precluded prospective clinical trials [
23
]. Current knowledge of therapy
is based on small clinical studies focused on chemotherapeutic approaches and clinical outcomes.
Cancers 2020,12, 2900 9 of 15
These studies have shown a role for anthracycline-containing chemotherapy regimens as ANKL
patients have shown a complete response [
20
]. Subsequently, a role for L-asparaginase in treating
ANKL was recognized. Treating ANKL cell lines with L-asparaginase results in apoptosis [
74
],
and L-asparginase has been included in various chemotherapy regimens to treat ANKL patients, such
as the SMILE (dexamethasone, methotrexate, ifosfamide, etoposide, and L-asparaginase), AspaMetDex
(L-asparaginase, methotrexate, and dexamethasone), or VIDL (etoposide, ifosfamide, dexamethasone,
and L-asparaginase) regimens, resulting in improved outcomes [
75
77
], However, no study has
compared these regimens in the setting of ANKL [
27
,
76
,
78
]. In addition, one study suggested that the
gemcitabine, cisplatin, and dexamethasone (GDP) regimen was ecacious in selected patients [
48
].
A complete response, including negativity for EBV DNA in the blood after treatment, is associated
with a better outcome, including overall survival [
76
]. However, prognosis remains poor. Patients who
achieve a complete remission (CR) after chemotherapy (including L-asparaginase) rarely survive more
than one year without further treatment.
11.2. Allogeneic Hematopoietic Cell Transplantation in ANKL
Allogeneic hematopoietic stem cell transplantation (SCT) improves outcome in ANKL patients.
In a study of eight patients who did not achieve complete remission (CR) before allogeneis SCT,
four patients reached CR, and two survived for several years [
75
]. Subsequent studies also have shown
the significant ecacy of allogeneic SCT for ANKL patients [
22
,
76
]. A total of 21 ANKL patients
enrolled in the International Bone Marrow Transplantation Registry (IBMTR) database underwent
allogeneic SCT, with most receiving L-asparaginase-containing chemotherapy before proceeding to
transplant [
79
]. Patients with a CR prior to allogeneic SCT showed a significantly better survival after
two years than those without a CR (38 vs. 0%) [
79
]. This study demonstrated that transplant can
provide durable disease control in a subset of ANKL patients who achieve CR before transplantation.
However, 76% of all patients died in the long run, mostly due to ANKL [
79
]. In summary, allogeneic
SCT in the setting ANKL might help extend survival in some patients, but success appears rather
limited, and novel therapies are needed for ANKL patients.
11.3. Novel Therapeutic Applications
Vulnerability of NK cells to targeted therapies could be better assessed through RNA sequencing
and drug sensitivity profiling of normal NK cells, as well as cells derived from NK cell neoplasms,
opening the gate to the practice of precision medicine in ANKL patients.
11.3.1. BCL2 Inhibitors
Dufva et al. investigated drug sensitivity profiling of nine NK cell lines, including three ANKL
and two ENKTL cell lines [
62
]. They concluded that NK cells show particular sensitivity to inhibition
of IL2–JAK–STAT signaling compared with other hematopoietic cells. The authors also reported high
ecacy of the JAK inhibitor ruxolitinib and the BCL2 family inhibitor navitoclax across all cell lines
(neoplastic and non-neoplastic) [
62
]. In addition, they observed that the BCL2 inhibitor venetoclax
and mTOR inhibitors were eective only against malignant NK cell lines [
62
]. Venetoclax was less
eective as a single agent compared with the more broad-spectrum BCL2 family inhibitor navitoclax,
but venetoclax exerted a synergistic eect with ruxolitinib more consistently and across all cell lines,
especially with the addition of the Aurora kinase (AURK) inhibitor alisertib [
62
]. In keeping with these
findings, El Hussein et al. demonstrated BCL2 protein overexpression in six of eight ANKL cases,
underscoring potential therapeutic applicability of BCL2 inhibitors in clinical settings [19].
11.3.2. Heat Shock Protein 90 (HSP90) Inhibitors
Other eective drug classes reported by Dufva et al. were heat shock protein 90 (HSP90)
inhibitors, Polo-like kinase (PLK) and cyclin-dependent kinase inhibitors, as well as histone deacetylase
inhibitors [
62
]. On the other hand, most NK cell lines were resistant to MEK inhibitors [
62
].
Cancers 2020,12, 2900 10 of 15
Glucocorticoids were highly eective against healthy NK cells compared to other cell types, but induced
responses only in few cell lines, implying glucocorticoid resistance in a subset of malignant NK cells [
62
].
Consistent with the role of HSP90 in JAK/STAT regulation [
80
], JAK and HSP90 inhibitors as well as
the Nedd8 activating enzyme (NAE) inhibitor pevonedistat were more eective in IL-2 stimulation than
resting NK cells, suggesting that the sensitivity of NK cells to JAK and HSP90 inhibition results largely
from inhibition of IL-2-derived JAK/STAT activation.
The role of the polycomb repressive complex 2 (PRC2) has been elucidated in the biology of T and
NK cell lymphomas [
81
84
]. PRC2 is a multiprotein complex composed of several core components
including EZH2, a histone methyltransferase responsible for epigenetic silencing of target genes via
trimethylation of H3K27. Increased or unchecked PRC2 activity hypermethylates H3K27, leading to
repression of tumor suppressor genes [
44
,
85
]. In one study, EZH2 and its’ eector H3K27me3 were
shown to be overexpressed by IHC in all three EBV-negative ANKL patients [
44
]. As we have discussed
above, activation of the JAK/STAT pathway is suggested to play a major role in overexpression of
MYC [
63
]. MYC, in turn, has been shown to interact with PRC2 through EZH2 and other cofactors such
as SUZ12/EED, and is responsible for inducing histone modification of H3K27me3 [
86
88
]. This axis
potentially contributes to the pathogenesis of NK cell neoplasms in general, and ANKL in particular.
This observation also provides new insights into potential druggable targets in ANKL through the
utilization of EZH2 inhibitors.
11.3.3. Other Approaches
Consideration of specific targeted therapies using ex vivo drug screening on patient-derived
xenografts might oer another approach for experimental therapies in patient with ANKL. Such an
approach has shown promise in patients with T cell neoplasms [
89
]. One example might be targeting
CD38 with daratumumab, as CD38 has already been shown to be expressed in ANKL [12,20].
12. Conclusions
Although the outcome of patients with ANKL is poor, and our understanding of ANKL has
evolved only slowly over time, substantial recent progress has been made in deciphering the molecular
alterations orchestrating the oncogenesis of ANKL. Pharmacologic experiments with therapeutic
agents tailored to optimize attack on leukemic NK cells and target recently characterized pathways in
ANKL, such as JAK/STAT, hold promise for the future. Implementing the data collected from anecdotal
observations into clinical trials, coupled with the creation of a multi-institutional genomic bank of
ANKL samples that can be analyzed by high throughput molecular methods, represents steps on the
journey towards understanding the biology of and designing eective therapies for this disease.
Funding: This research received no external funding.
Conflicts of Interest: The authors declare no conflict of interest.
References
1.
Chan, J.K.C.; Jae, E.S.; Ko, Y.H. Aggressive NK-Cell Leukaemia. In WHO Classification of Tumours of
Haematopoietic and Lymphoid Tissues; IARC Press: Lyon, France, 2017; pp. 353–354.
2.
Choi, Y.L.; Makishima, H.; Ohashi, J.; Yamashita, Y.; Ohki, R.; Koinuma, K.; Ota, J.; Isobe, Y.; Ishida, F.;
Oshimi, K.; et al. DNA microarray analysis of natural killer cell-type lymphoproliferative disease of granular
lymphocytes with purified CD3-CD56+fractions. Leukemia 2004,18, 556–565. [PubMed]
3.
Epling-Burnette, P.K.; Bai, F.; Wei, S.; Chaurasia, P.; Painter, J.S.; Olashaw, N.; Hamilton, A.; Sebti, S.; Djeu, J.Y.;
Loughran, T.P. ERK couples chronic survival of NK cells to constitutively activated Ras in lymphoproliferative
disease of granular lymphocytes (LDGL). Oncogene 2004,23, 9220–9229. [CrossRef] [PubMed]
4.
Scquizzato, E.; Teramo, A.; Miorin, M.; Facco, M.; Piazza, F.; Noventa, F.; Trentin, L.; Agostini, C.; Zambello, R.;
Semenzato, G. Genotypic evaluation of killer immunoglobulin-like receptors in NK-type lymphoproliferative
disease of granular lymphocytes. Leukemia 2007,21, 1060–1069. [CrossRef] [PubMed]
Cancers 2020,12, 2900 11 of 15
5.
Zambello, R.; Falco, M.; Della Chiesa, M.; Trentin, L.; Carollo, D.; Castriconi, R.; Cannas, G.; Carlomagno, S.;
Cabrelle, A.; Lamy, T.; et al. Expression and function of KIR and natural cytotoxicity receptors in NK-type
lymphoproliferative diseases of granular lymphocytes. Blood 2003,102, 1797–1805. [PubMed]
6.
Fernandez, L.A.; Pope, B.; Lee, C.; Zayed, E. Aggressive natural killer cell leukemia in an adult with
establishment of an NK cell line. Blood 1986,67, 925–930.
7.
Lmamura, N.; Kusunoki, Y.; Kajihara, H.; Okada, K.; Kuramoto, A. Aggressive Natural Killer Cell
Leukemia/Lymphoma with N901-Positive: Surface Phenotype: Evidence for the Existence of a Third
Lineage in Lymphoid Cells. Acta Haematol. 1988,80, 121–128. [CrossRef]
8.
Koizumi, S.; Seki, H.; Tachinami, T.; Taniguchi, M.; Matsuda, A.; Taga, K.; Nakarai, T.; Kato, E.; Taniguchi, N.;
Nakamura, H. Malignant clonal expansion of large granular lymphocytes with a Leu-11+, Leu-7- surface
phenotype:
In vitro
responsiveness of malignant cells to recombinant human interleukin 2. Blood
1986
,68,
1065–1073.
9.
Kotani, H.; Ishikura, H.; Hayashi, H.; Tsunematsu, T. Aggressive LGL leukemia associated with recurrent
fever and gastrointestinal ulcer. Acta Haematol. Jpn. 1988,51, 272.
10. Ohno, Y.; Kamezaki, H.A.; Imanaka, H.; Takahashi, T.; Arita, Y.; Amakawa, H.; Uchiyama, R.; Fukuhara, T.;
Kita, S. T3-, Leull+, Leu7- LGL leukemia with germline configuration of TcR gene. Acta Haematol. Jpn.
1987
,
50, 338.
11.
Takiguchi, T.; Yoshioka, R.; Tachibana, J.; Maekawa, H.; Hirose, Y.; Konda, S.; Konishi, F. NK cell leukemia
and T-y lymphocytosis. In New Diagnostic Technique for Leukemia; Ishiyake Publishers: Tokyo, Japan, 1987;
pp. 89–99.
12.
Imamura, N.; Kusunoki, Y.; Kawa-Ha, K.; Yumura, K.; Hara, J.; Oda, K.; Abe, K.; Dohy, H.; Inada, T.;
Kajihara, H.; et al. Aggressive natural killer cell leukaemia/lymphoma: Report of four cases and review of
the literature Possible Existence of a New Clinical Entity Originating From the Third Lineage of Lymphoid
Cells. Br. J. Haematol. 1990,75, 49–56. [CrossRef]
13.
Soler, J.; Bordes, R.; Ortouno, F.; Montagud, M.; Martorell, J.; Pons, C.; Nomdedeu, J.; Lopez-Lopez, J.J.;
Prat, J.; Rutllant, M. Aggressive natural killer cell leukaemia/lymphoma in two patients with lethal midline
granuloma. Br. J. Haematol. 1994,86, 659–662. [CrossRef] [PubMed]
14.
Mori, N.; Yamashita, Y.; Tsuzuki, T.; Nakayama, A.; Nakazawa, M.; Hasegawa, Y.; Kojima, H.;
Nagasawa, T. Lymphomatous features of aggressive NK cell leukaemia/lymphoma with massive necrosis,
haemophagocytosis and EB virus infection. Histopathology 2000,37, 363–371. [CrossRef] [PubMed]
15.
Sun, T.; Brody, J.; Susin, M.; Teichberg, S.; Koduru, P.; Urmacher, C.; Hajdu, S.I.; Marino, J.; Hall, W.W.
Aggressive Natural Killer Cell Lymphoma/Leukemia: A recently recognized clinicopathologic entity. Am. J.
Surg. Pathol. 1993,17, 1289–1299. [CrossRef] [PubMed]
16.
Quintanilla-Martinez, L.; Jae, E.S. Commentary: Aggressive NK cell lymphomas: Insights into the spectrum
of NK cell derived malignancies. Histopathology 2000,37, 372–374. [CrossRef] [PubMed]
17.
Chan, J.K.C.; Jae, E.; Ralfkiaer, E.; Ko, Y.H. Aggressive NK-Cell Leukemia. In World Health Organization
Classification of Tumors, Tumors of Haematopoietic and Lymphoid Tissues; IARC Press: Lyon, France, 2008;
pp. 197–200.
18.
Harris, N.L.; Jae, E.S.; Stein, H.; Banks, P.M.; Chan, J.K.; Cleary, M.L.; Delsol, G.; De Wolf-Peeters, C.;
Falini, B.; Gatter, K.C.; et al. A revised European-American classification of lymphoid neoplasms: A proposal
from the International Lymphoma Study Group. Blood 1994,84, 1361–1392. [CrossRef] [PubMed]
19.
El Hussein, S.; Patel, K.P.; Fang, H.; Thakral, B.; Loghavi, S.; Kanagal-Shamanna, R.; Konoplev, S.; Jabbour, E.J.;
Medeiros, L.J.; Khoury, J.D. Genomic and Immunophenotypic Landscape of Aggressive NK-Cell Leukemia.
Am. J. Surg. Pathol. 2020. [CrossRef] [PubMed]
20.
Suzuki, R.; Suzumiya, J.; Nakamura, S.; Aoki, S.; Notoya, A.; Ozaki, S.; Gondo, H.; Hino, N.; Mori, H.;
Sugimori, H.; et al. Aggressive natural killer-cell leukemia revisited: Large granular lymphocyte leukemia of
cytotoxic NK cells. Leukemia 2004,18, 763–770. [CrossRef]
21.
Song, S.-Y.; Kim, W.S.; Ko, Y.H.; Kim, K.; Lee, M.H.; Park, K. Aggressive natural killer cell leukemia: Clinical
features and treatment outcome. Haematologica 2002,87, 1343–1345.
22.
Tang, Y.-T.; Wang, D.; Luo, H.; Xiao, M.; Zhou, H.-S.; Liu, D.; Ling, S.-P.; Wang, N.; Hu, X.-L.; Luo, Y.; et al.
Aggressive NK-cell leukemia: Clinical subtypes, molecular features, and treatment outcomes. Blood Cancer J.
2017,7, 660. [CrossRef]
23. Ishida, F. Aggressive NK-Cell Leukemia. Front. Pediatr. 2018,6, 292. [CrossRef]
Cancers 2020,12, 2900 12 of 15
24.
Ruskova, A.; Thula, R.; Chan, G. Aggressive Natural Killer-Cell Leukemia: Report of five cases and review
of the literature. Leuk. Lymphoma 2004,45, 2427–2438. [CrossRef] [PubMed]
25.
Ryder, J.; Wang, X.; Bao, L.; Gross, S.A.; Hua, F.; Irons, R.D. Aggressive natural killer cell leukemia: Report of
a Chinese series and review of the literature. Int. J. Hematol. 2007,85, 18–25. [CrossRef]
26.
Zhang, Q.; Jing, W.; Ouyang, J.; Zeng, H.; George, S.K.; Liu, Z. Six cases of aggressive natural killer-cell
leukemia in a Chinese population. Int. J. Clin. Exp. Pathol. 2014,7, 3423–3431. [PubMed]
27.
Li, C.; Tian, Y.; Wang, J.; Zhu, L.; Huang, L.; Wang, N.; Xu, D.; Cao, Y.; Li, J.; Zhou, J. Abnormal
immunophenotype provides a key diagnostic marker: A report of 29 cases of de novo aggressive natural
killer cell leukemia. Transl. Res. 2014,163, 565–577. [CrossRef] [PubMed]
28.
Luetke-Eversloh, M.; Killig, M.; Romagnani, C. Signatures of human NK cell development and terminal
dierentiation. Front. Immunol. 2013,4, 499. [CrossRef] [PubMed]
29.
Poli, A.; Michel, T.; Theresine, M.; Andres, E.; Hentges, F.; Zimmer, J. CD56bright natural killer (NK) cells:
An important NK cell subset. Immunology 2009,126, 458–465. [CrossRef] [PubMed]
30.
Jiang, N.G.; Jin, Y.M.; Niu, Q.; Zeng, T.T.; Su, J.; Zhu, H.L. Flow cytometric immunophenotyping is of
great value to diagnosis of natural killer cell neoplasms involving bone marrow and peripheral blood.
Ann. Hematol. 2013,92, 89–96. [CrossRef]
31.
Kurt, H.; Jorgensen, J.L.; Amin, H.M.; Patel, K.P.; Wang, S.A.; Lin, P.; Kanagal-Shamanna, R.; Loghavi, S.;
Thakral, B.; Khogeer, H.A.; et al. Chronic lymphoproliferative disorder of NK-cells: A single-institution
review with emphasis on relative utility of multimodality diagnostic tools. Eur. J. Haematol.
2018
,100,
444–454. [CrossRef]
32.
Borrego, F.; Masilamani, M.; Kabat, J.; Sanni, T.B.; Coligan, J.E. The cell biology of the human natural killer
cell CD94/NKG2A inhibitory receptor. Mol. Immunol. 2005,42, 485–488. [CrossRef]
33.
Braud, V.M.; Allan, D.S.; O’Callaghan, C.A.; Soderstrom, K.; D’Andrea, A.; Ogg, G.S.; Lazetic, S.; Young, N.T.;
Bell, J.I.; Phillips, J.H.; et al. HLA-E binds to natural killer cell receptors CD94/NKG2A, B and C. Nature
1998
,
391, 795–799. [CrossRef]
34.
Arlettaz, L.; Villard, J.; de Rham, C.; Degermann, S.; Chapuis, B.; Huard, B.; Roosnek, E. Activating
CD94:NKG2C and inhibitory CD94:NKG2A receptors are expressed by distinct subsets of committed CD8+
TCR alphabeta lymphocytes. Eur. J. Immunol. 2004,34, 3456–3464. [CrossRef] [PubMed]
35.
Cantoni, C.; Biassoni, R.; Pende, D.; Sivori, S.; Accame, L.; Pareti, L.; Semenzato, G.; Moretta, L.; Moretta, A.;
Bottino, C. The activating form of CD94 receptor complex: CD94 covalently associates with the Kp39 protein
that represents the product of the NKG2-C gene. Eur. J. Immunol. 1998,28, 327–338. [CrossRef]
36.
Lima, M.; Spinola, A.; Fonseca, S.; Santos, A.H.; Rodrigues, J.; Oliveira, L.; Queiros, M.L.; Santos, M.;
Goncalves, M.; Lau, C.; et al. Aggressive mature natural killer cell neoplasms: Report on a series of
12 European patients with emphasis on flow cytometry based immunophenotype and DNA content of
neoplastic natural killer cells. Leuk. Lymphoma 2015,56, 103–112. [CrossRef] [PubMed]
37.
Ogata, M.; Imamura, T.; Mizunoe, S.; Ohtsuka, E.; Kikuchi, H.; Nasu, M. Natural killer cell-type body
cavity lymphoma following chronic active Epstein-Barr virus infection. Am. J. Hematol.
2003
,73, 126–130.
[CrossRef]
38.
Lima, M.; Goncalves, C.; Teixeira, M.A.; Franca, M.; Canelhas, A.; Pina, R.; Lopes, V.; Queiros, M.L.;
Fonseca, S.; Santos, A.H.; et al. Aggressive natural-killer cell lymphoma presenting with skin lesions, breast
nodule, suprarenal masses and life-threatening pericardial and pleural eusions. Leuk. Lymphoma
2001
,42,
1385–1391. [CrossRef]
39.
Pullarkat, V.A.; Medeiros, L.J.; Brynes, R.K. Body cavity-based presentation of natural killer cell lymphoma.
Leuk. Lymphoma 2005,46, 293–296. [CrossRef]
40.
Lepeak, L.M.; Yang, D.T.; Chang, J.E. Extranodal NK/T-cell lymphoma presenting with primary cardiac
involvement. Hematol. Rep. 2011,3, e9. [CrossRef]
41.
Kawa-Ha, K.; Ishihara, S.; Ninomiya, T.; Yumura-Yagi, K.; Hara, J.; Murayama, F.; Tawa, A.; Hirai, K.
CD3-negative lymphoproliferative disease of granular lymphocytes containing Epstein-Barr viral DNA.
J. Clin. Investig. 1989,84, 51–55. [CrossRef]
42.
Kanegane, H.; Yachie, A.; Miyawaki, T.; Tosato, G. EBV-NK cells interactions and lymphoproliferative
disorders. Leuk Lymphoma 1998,29, 491–498. [CrossRef]
43.
Ng, S.B.; Khoury, J.D. Epstein-Barr virus in lymphoproliferative processes: An update for the diagnostic
pathologist. Adv. Anat. Pathol. 2009,16, 40–55. [CrossRef]
Cancers 2020,12, 2900 13 of 15
44.
Gao, J.; Behdad, A.; Ji, P.; Wolniak, K.L.; Frankfurt, O.; Chen, Y.H. EBV-negative aggressive NK-cell
leukemia/lymphoma: A clinical and pathological study from a single institution. Mod. Pathol.
2017
,30,
1100–1115. [CrossRef] [PubMed]
45.
Nicolae, A.; Ganapathi, K.A.; Pham, T.H.; Xi, L.; Torres-Cabala, C.A.; Nanaji, N.M.; Zha, H.D.; Fan, Z.;
Irwin, S.; Pittaluga, S.; et al. EBV-negative Aggressive NK-cell Leukemia/Lymphoma: Clinical, Pathologic,
and Genetic Features. Am. J. Surg. Pathol. 2017,41, 67–74. [CrossRef] [PubMed]
46.
Jitani, A.K.; Khonglah, Y.; Kumar, R.; Gogoi, B.B.; Jajodia, E. Natural Killer Cell Lymphoma: A Case with
Classification Dilemma. J. Clin. Diagn. Res. 2016,10, ED07. [CrossRef] [PubMed]
47.
Matano, S.; Nakamura, S.; Nakamura, S.; Annen, Y.; Hattori, N.; Kobayashi, K.; Kyoda, K.; Sugimoto, T.
Monomorphic agranular natural killer cell lymphoma/leukemia with no Epstein-Barr virus association.
Acta Haematol. 1999,101, 206–208. [CrossRef]
48.
Zhang, H.; Meng, Q.; Yin, W.; Xu, L.; Lie, L. Adult aggressive natural killer cell leukemia. Am. J. Med. Sci.
2013,346, 56–63. [CrossRef]
49.
Park, J.A.; Jun, K.R.; Nam, S.H.; Ghim, T.T. Favorable outcome in a child with EBV-negative aggressive NK
cell leukemia. Int. J. Hematol. 2013,97, 673–676. [CrossRef]
50.
Ko, Y.H.; Park, S.; Kim, K.; Kim, S.J.; Kim, W.S. Aggressive natural killer cell leukemia: Is Epstein-Barr virus
negativity an indicator of a favorable prognosis? Acta Haematol. 2008,120, 199–206. [CrossRef]
51.
Wong, K.F.; Chan, J.K.; Kwong, Y.L. Identification of del (6) (q21q25) as a recurring chromosomal abnormality
in putative NK cell lymphoma/leukaemia. Br. J. Haematol. 1997,98, 922–926. [CrossRef]
52.
Yang, C.F.; Hsu, C.Y.; Ho, D.M. Aggressive natural killer (NK)-cell leukaemia and extranodal NK/T-cell
lymphoma are two distinct diseases that dier in their clinical presentation and cytogenetic findings.
Histopathology 2018,72, 955–964. [CrossRef]
53.
Nakashima, Y.; Tagawa, H.; Suzuki, R.; Karnan, S.; Karube, K.; Ohshima, K.; Muta, K.; Nawata, H.;
Morishima, Y.; Nakamura, S.; et al. Genome-wide array-based comparative genomic hybridization of natural
killer cell lymphoma/leukemia: Dierent genomic alteration patterns of aggressive NK-cell leukemia and
extranodal Nk/T-cell lymphoma, nasal type. Genes Chromosom. Cancer 2005,44, 247–255. [CrossRef]
54.
Iqbal, J.; Kucuk, C.; Deleeuw, R.J.; Srivastava, G.; Tam, W.; Geng, H.; Klinkebiel, D.; Christman, J.K.; Patel, K.;
Cao, K.; et al. Genomic analyses reveal global functional alterations that promote tumor growth and
novel tumor suppressor genes in natural killer-cell malignancies. Leukemia
2009
,23, 1139–1151. [CrossRef]
[PubMed]
55.
Kucuk, C.; Jiang, B.; Hu, X.; Zhang, W.; Chan, J.K.; Xiao, W.; Lack, N.; Alkan, C.; Williams, J.C.; Avery, K.N.; et al.
Activating mutations of STAT5B and STAT3 in lymphomas derived from gammadelta-T or NK cells.
Nat. Commun. 2015,6, 6025. [CrossRef] [PubMed]
56.
Lee, S.; Park, H.Y.; Kang, S.Y.; Kim, S.J.; Hwang, J.; Lee, S.; Kwak, S.H.; Park, K.S.; Yoo, H.Y.; Kim, W.S.; et al.
Genetic alterations of JAK/STAT cascade and histone modification in extranodal NK/T-cell lymphoma nasal
type. Oncotarget 2015,6, 17764–17776. [CrossRef]
57.
Koo, G.C.; Tan, S.Y.; Tang, T.; Poon, S.L.; Allen, G.E.; Tan, L.; Chong, S.C.; Ong, W.S.; Tay, K.; Tao, M.; et al.
Janus kinase 3-activating mutations identified in natural killer/T-cell lymphoma. Cancer Discov.
2012
,2,
591–597. [CrossRef] [PubMed]
58.
Bouchekioua, A.; Scourzic, L.; de Wever, O.; Zhang, Y.; Cervera, P.; Aline-Fardin, A.; Mercher, T.; Gaulard, P.;
Nyga, R.; Jeziorowska, D.; et al. JAK3 deregulation by activating mutations confers invasive growth
advantage in extranodal nasal-type natural killer cell lymphoma. Leukemia 2014,28, 338–348. [CrossRef]
59.
Huang, Y.; de Reynies, A.; de Leval, L.; Ghazi, B.; Martin-Garcia, N.; Travert, M.; Bosq, J.; Briere, J.; Petit, B.;
Thomas, E.; et al. Gene expression profiling identifies emerging oncogenic pathways operating in extranodal
NK/T-cell lymphoma, nasal type. Blood 2010,115, 1226–1237. [CrossRef]
60.
Karube, K.; Nakagawa, M.; Tsuzuki, S.; Takeuchi, I.; Honma, K.; Nakashima, Y.; Shimizu, N.; Ko, Y.H.;
Morishima, Y.; Ohshima, K.; et al. Identification of FOXO3 and PRDM1 as tumor-suppressor gene candidates
in NK-cell neoplasms by genomic and functional analyses. Blood 2011,118, 3195–3204. [CrossRef]
61.
Jiang, L.; Gu, Z.H.; Yan, Z.X.; Zhao, X.; Xie, Y.Y.; Zhang, Z.G.; Pan, C.M.; Hu, Y.; Cai, C.P.; Dong, Y.; et al.
Exome sequencing identifies somatic mutations of DDX3X in natural killer/T-cell lymphoma. Nat. Genet.
2015,47, 1061–1066. [CrossRef]
Cancers 2020,12, 2900 14 of 15
62.
Dufva, O.; Kankainen, M.; Kelkka, T.; Sekiguchi, N.; Awad, S.A.; Eldfors, S.; Yadav, B.; Kuusanmaki, H.;
Malani, D.; Andersson, E.I.; et al. Aggressive natural killer-cell leukemia mutational landscape and drug
profiling highlight JAK-STAT signaling as therapeutic target. Nat. Commun. 2018,9, 1567. [CrossRef]
63.
Huang, L.; Liu, D.; Wang, N.; Ling, S.; Tang, Y.; Wu, J.; Hao, L.; Luo, H.; Hu, X.; Sheng, L.; et al. Integrated
genomic analysis identifies deregulated JAK/STAT-MYC-biosynthesis axis in aggressive NK-cell leukemia.
Cell Res. 2018,28, 172–186. [CrossRef]
64.
Villarino, A.V.; Kanno, Y.; Ferdinand, J.R.; O’Shea, J.J. Mechanisms of Jak/STAT signaling in immunity and
disease. J. Immunol. 2015,194, 21–27. [CrossRef] [PubMed]
65.
Brooks, A.J.; Dai, W.; O’Mara, M.L.; Abankwa, D.; Chhabra, Y.; Pelekanos, R.A.; Gardon, O.; Tunny, K.A.;
Blucher, K.M.; Morton, C.J.; et al. Mechanism of activation of protein kinase JAK2 by the growth hormone
receptor. Science 2014,344, 1249783. [CrossRef] [PubMed]
66.
Langlais, D.; Couture, C.; Balsalobre, A.; Drouin, J. The Stat3/GR interaction code: Predictive value of
direct/indirect DNA recruitment for transcription outcome. Mol. Cell
2012
,47, 38–49. [CrossRef] [PubMed]
67.
Walker, S.R.; Nelson, E.A.; Yeh, J.E.; Pinello, L.; Yuan, G.C.; Frank, D.A. STAT5 outcompetes STAT3 to
regulate the expression of the oncogenic transcriptional modulator BCL6. Mol. Cell Biol.
2013
,33, 2879–2890.
[CrossRef]
68.
Wan, C.K.; Oh, J.; Li, P.; West, E.E.; Wong, E.A.; Andraski, A.B.; Spolski, R.; Yu, Z.X.; He, J.; Kelsall, B.L.; et al.
The cytokines IL-21 and GM-CSF have opposing regulatory roles in the apoptosis of conventional dendritic
cells. Immunity 2013,38, 514–527. [CrossRef]
69.
Yang, X.P.; Ghoreschi, K.; Steward-Tharp, S.M.; Rodriguez-Canales, J.; Zhu, J.; Grainger, J.R.; Hirahara, K.;
Sun, H.W.; Wei, L.; Vahedi, G.; et al. Opposing regulation of the locus encoding IL-17 through direct,
reciprocal actions of STAT3 and STAT5. Nat. Immunol. 2011,12, 247–254. [CrossRef]
70.
Yu, H.; Pardoll, D.; Jove, R. STATs in cancer inflammation and immunity: A leading role for STAT3.
Nat. Rev. Cancer 2009,9, 798–809. [CrossRef]
71.
Dobashi, A.; Tsuyama, N.; Asaka, R.; Togashi, Y.; Ueda, K.; Sakata, S.; Baba, S.; Sakamoto, K.;
Hatake, K.; Takeuchi, K. Frequent BCOR aberrations in extranodal NK/T-Cell lymphoma, nasal type.
Genes Chromosom. Cancer 2016,55, 460–471. [CrossRef]
72.
Kwong, Y.L.; Chan, T.S.Y.; Tan, D.; Kim, S.J.; Poon, L.M.; Mow, B.; Khong, P.L.; Loong, F.; Au-Yeung, R.;
Iqbal, J.; et al. PD1 blockade with pembrolizumab is highly eective in relapsed or refractory NK/T-cell
lymphoma failing l-asparaginase. Blood 2017,129, 2437–2442. [CrossRef]
73.
Green, M.R.; Rodig, S.; Juszczynski, P.; Ouyang, J.; Sinha, P.; O’Donnell, E.; Neuberg, D.; Shipp, M.A.
Constitutive AP-1 activity and EBV infection induce PD-L1 in Hodgkin lymphomas and posttransplant
lymphoproliferative disorders: Implications for targeted therapy. Clin. Cancer Res.
2012
,18, 1611–1618.
[CrossRef]
74.
Ando, M.; Sugimoto, K.; Kitoh, T.; Sasaki, M.; Mukai, K.; Ando, J.; Egashira, M.; Schuster, S.M.; Oshimi, K.
Selective apoptosis of natural killer-cell tumours by l-asparaginase. Br. J. Haematol.
2005
,130, 860–868.
[CrossRef] [PubMed]
75.
Ishida, F.; Ko, Y.H.; Kim, W.S.; Suzumiya, J.; Isobe, Y.; Oshimi, K.; Nakamura, S.; Suzuki, R. Aggressive
natural killer cell leukemia: Therapeutic potential of L-asparaginase and allogeneic hematopoietic stem cell
transplantation. Cancer Sci. 2012,103, 1079–1083. [CrossRef] [PubMed]
76.
Jung, K.S.; Cho, S.H.; Kim, S.J.; Ko, Y.H.; Kang, E.S.; Kim, W.S. L-asparaginase-based regimens followed
by allogeneic hematopoietic stem cell transplantation improve outcomes in aggressive natural killer cell
leukemia. J. Hematol. Oncol. 2016,9, 41. [CrossRef] [PubMed]
77.
Gao, L.M.; Zhao, S.; Liu, W.P.; Zhang, W.Y.; Li, G.D.; Kucuk, C.; Hu, X.Z.; Chan, W.C.; Tang, Y.; Ding, W.S.; et al.
Clinicopathologic Characterization of Aggressive Natural Killer Cell Leukemia Involving Dierent Tissue
Sites. Am. J. Surg. Pathol. 2016,40, 836–846. [CrossRef] [PubMed]
78.
Ito, T.; Makishima, H.; Nakazawa, H.; Kobayashi, H.; Shimodaira, S.; Nakazawa, Y.; Kitano, K.; Matsuda, K.;
Hidaka, E.; Ishida, F. Promising approach for aggressive NK cell leukaemia with allogeneic haematopoietic
cell transplantation. Eur. J. Haematol. 2008,81, 107–111. [CrossRef] [PubMed]
79.
Hamadani, M.; Kanate, A.S.; DiGilio, A.; Ahn, K.W.; Smith, S.M.; Lee, J.W.; Ayala, E.; Chao, N.; Hari, P.;
Bolanos-Meade, J.; et al. Allogeneic Hematopoietic Cell Transplantation for Aggressive NK Cell Leukemia.
A Center for International Blood and Marrow Transplant Research Analysis. Biol. Blood Marrow Transplant.
2017,23, 853–856. [CrossRef]
Cancers 2020,12, 2900 15 of 15
80.
Marubayashi, S.; Koppikar, P.; Taldone, T.; Abdel-Wahab, O.; West, N.; Bhagwat, N.; Caldas-Lopes, E.;
Ross, K.N.; Gonen, M.; Gozman, A.; et al. HSP90 is a therapeutic target in JAK2-dependent myeloproliferative
neoplasms in mice and humans. J. Clin. Investig. 2010,120, 3578–3593. [CrossRef]
81.
Nagel, S.; Venturini, L.; Marquez, V.E.; Meyer, C.; Kaufmann, M.; Scherr, M.; MacLeod, R.A.; Drexler, H.G.
Polycomb repressor complex 2 regulates HOXA9 and HOXA10, activating ID2 in NK/T-cell lines. Mol. Cancer
2010,9, 151. [CrossRef]
82.
Yan, J.; Ng, S.B.; Tay, J.L.; Lin, B.; Koh, T.L.; Tan, J.; Selvarajan, V.; Liu, S.C.; Bi, C.; Wang, S.; et al. EZH2
overexpression in natural killer/T-cell lymphoma confers growth advantage independently of histone
methyltransferase activity. Blood 2013,121, 4512–4520. [CrossRef]
83.
Yan, J.; Li, B.; Lin, B.; Lee, P.T.; Chung, T.H.; Tan, J.; Bi, C.; Lee, X.T.; Selvarajan, V.; Ng, S.B.; et al. EZH2
phosphorylation by JAK3 mediates a switch to noncanonical function in natural killer/T-cell lymphoma.
Blood 2016,128, 948–958. [CrossRef]
84.
Kim, S.H.; Yang, W.I.; Min, Y.H.; Ko, Y.H.; Yoon, S.O. The role of the polycomb repressive complex pathway
in T and NK cell lymphoma: Biological and prognostic implications. Tumour. Biol.
2016
,37, 2037–2047.
[CrossRef] [PubMed]
85.
Laugesen, A.; Hojfeldt, J.W.; Helin, K. Role of the Polycomb Repressive Complex 2 (PRC2) in Transcriptional
Regulation and Cancer. Cold Spring Harb. Perspect. Med. 2016,6. [CrossRef] [PubMed]
86.
Sander, S.; Bullinger, L.; Klapproth, K.; Fiedler, K.; Kestler, H.A.; Barth, T.F.; Moller, P.; Stilgenbauer, S.;
Pollack, J.R.; Wirth, T. MYC stimulates EZH2 expression by repression of its negative regulator miR-26a.
Blood 2008,112, 4202–4212. [CrossRef] [PubMed]
87.
Benetatos, L.; Vartholomatos, G.; Hatzimichael, E. Polycomb group proteins and MYC: The cancer connection.
Cell Mol. Life Sci. 2014,71, 257–269. [CrossRef]
88.
Neri, F.; Zippo, A.; Krepelova, A.; Cherubini, A.; Rocchigiani, M.; Oliviero, S. Myc regulates the transcription
of the PRC2 gene to control the expression of developmental genes in embryonic stem cells. Mol. Cell Biol.
2012,32, 840–851. [CrossRef]
89.
Wu, C.H.; Yang, C.Y.; Wang, L.; Gao, H.X.; Rakhshandehroo, T.; Afghani, S.; Pincus, L.; Balassanian, R.;
Rubenstein, J.; Gill, R.; et al. Cutaneous T cell lymphoma PDX drug screening platform identifies cooperation
between inhibitions of PI3Kalpha/delta and HDAC. J. Investig. Dermatol. 2020. [CrossRef]
©
2020 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access
article distributed under the terms and conditions of the Creative Commons Attribution
(CC BY) license (http://creativecommons.org/licenses/by/4.0/).
... [4][5][6] The diagnosis can be made when CD56-positive NK cells, appearing as large granular lymphocytes with azurophilic granules, present leukemic manifestations in peripheral blood and bone marrow (BM). [7] There is a strong association with Epstein-Barr virus (EBV) infection. [8] Extranodal NK/T-cell lymphoma (ENKTL) is an EBV-associated malignant neoplasm characterized by extranodal proliferation of NK or T cells. ...
... JAK-STAT pathway mutations have been reported in 21.0-66.6% of ANKL cases and in approximately 20.0% of ENKTL cases. [7,31,[39][40][41][49][50][51] In our study, these mutations were identi ed with frequencies of 60.0% in both ANKL and ENKTL-BM. JAK-STAT pathway mutations have also been reported in various types of T-cell leukemia/lymphomas. ...
... Cox regression analysis for candidate prognostic factors for ANKL Although numerous studies have attempted to genetically differentiate ANKL from ENKTL, no study has identi ed ANKL-speci c genetic characteristics.[7,21,29,31,[38][39][40][41] Moreover, no genetic prognostic factor has been suggested for ANKL.[7] ...
Preprint
Full-text available
Aggressive NK-cell leukemia (ANKL) shares common clinicopathological features with extranodal NK/T-cell lymphoma with bone marrow (BM) involvement (ENKTL-BM), making their distinction challenging in BM examination. Despite numerous studies, genetic differences between the two diseases remained largely unclear. To investigate the genetic and clinical differences between ANKL and ENKTL-BM, we performed targeted sequencing of 282 genes and survival analyses on 15 ANKL and 5 ENKTL-BM patients. Mutation frequency of FAT family genes was higher in ANKL than in ENKTL-BM (80.0% vs. 0.0%, P = 0.004), and FAT1 gene mutations were associated with significantly lower survival rates in ANKL patients ( P = 0.002). Copy number alterations including 11q loss and 4q loss were detected exclusively in ANKL. The interval from symptom onset to death was significantly shorter (113.0 vs. 440.5 days, P = 0.027) and survival rate was significantly lower ( P = 0.004) in ANKL than in ENKTL-BM. In conclusion, ANKL exhibited a higher mutation frequency of FAT family genes, a more acute fulminant clinical course, and worse prognosis than ENKTL-BM, indicating that ANKL and ENKTL-BM can be distinguished both genetically and clinically. We expect the identified FAT1 gene mutations to serve as novel prognostic factors for ANKL.
... Patients typically present with constitutional symptoms, hepatosplenomegaly, and complications such as hemophagocytic syndrome or disseminated intravascular coagulation [5]. The standard treatment involves L-asparaginase-based chemotherapy followed by allogeneic hematopoietic stem cell transplantation (allo-HSCT) [4]. ...
... However, some alternative treatments like cisplatin-based chemotherapy have shown promise in achieving complete remission in isolated cases [7]. Recent molecular studies have identified potential therapeutic targets, including activation of the JAK/ STAT pathway, epigenetic dysregulation, and impaired TP53 function, which could pave the way for novel treatment strategies [5]. Nonetheless, these treatments often fail to secure long-term survival, as patients frequently relapse. ...
Article
Full-text available
Aggressive NK-cell leukemia (ANKL) is a rare and highly malignant lymphoproliferative disorder associated with poor prognosis and high mortality rates. The disease is closely linked to Epstein-Barr virus (EBV) and predominantly affects populations in Asia and Latin America. Allogeneic hematopoietic stem cell transplantation (allo-HSCT) has emerged as a promising treatment strategy, yet its efficacy and prognosis in ANKL patients remain to be systematically evaluated. We conducted a systematic review and meta-analysis by searching PubMed, Embase, Web of Science, and the Cochrane Library for studies published up to April 2025. The objective was to evaluate the outcomes of allo-HSCT in patients with ANKL. Six studies met the inclusion criteria. The primary outcomes included overall survival (OS) and progression-free survival (PFS), reported as hazard ratios (HRs), as well as standardized mean differences (SMDs) and median follow-up times for survivors (reported in months with ranges). Depending on the degree of heterogeneity—assessed using the I² statistic—random-effects or fixed-effects models were applied. Sensitivity analyses were conducted by sequentially excluding individual studies, and publication bias was assessed using funnel plots. Based on the analysis, a total of six studies comprising 295 ANKL patients who underwent allo-HSCT were included. The SMD for the median follow-up duration among survivors was 1.21 (95% CI: 0.39–2.02), indicating notable variation in follow-up times across studies. Heterogeneity for this outcome was minimal, with an I² of 0% and a p-value of 0.6621. Regarding the effect of allo-HSCT on overall survival, the pooled HR was 0.47 (95% CI: 0.32–0.68), demonstrating a statistically significant survival benefit associated with the procedure. Heterogeneity in this analysis was also low (I² = 0.0%, p = 0.684), reflecting strong consistency across the included studies. For PFS, the pooled HR was 0.22 (95% CI: 0.12–0.40), again indicating a significant improvement in outcomes. However, moderate heterogeneity was observed in this analysis (I² = 68.4%, p = 0.032). Funnel plot analysis revealed no significant evidence of publication bias, suggesting that the included data were robust and not substantially affected by selective reporting. This meta-analysis demonstrates that allo-HSCT significantly improves survival outcomes in patients with ANKL. The pooled hazard ratio of 0.47 indicates a favorable impact on overall survival. Additionally, the median survival time among survivors is notably longer, further supporting the efficacy of allo-HSCT. These results highlight the potential of allo-HSCT as a promising therapeutic strategy for ANKL, emphasizing the need for continued research to refine treatment protocols and enhance patient outcomes.
... The clinical course is frequently fulminant, rapidly progressing to disseminated intravascular coagulation (DIC) and HLH, resulting in multiorgan failure [6]. In terms of molecular characteristics, the molecular abnormalities observed in ANKL can be categorized into four primary groups: hyperactivation of the JAK/STAT pathway, epigenetic deregulation, dysfunction in TP53 and DNA repair mechanisms, and overexpression of PD-1/PD-L1 [7,8]. These genetic mutations are significantly correlated with a shorter survival duration. ...
Article
Full-text available
Aggressive NK-cell leukemia (ANKL) is a scarce mature NK-cell neoplasm frequently associated with Epstein-Barr virus (EBV) infection. We report the case of a 47-year-old male patient who was admitted to the hospital due to recurrent fever, jaundice, and dyspnea, and was diagnosed with ANKL accompanied by multi-organ failure. Upon transfer to the intensive care unit (ICU), he received emergency chemotherapy consisting of etoposide, pegaspargase, and gemcitabine, in conjunction with organ support therapies including DPMAS, LPE, and CVVHD. Subsequently, the patient’s condition stabilized, and he was discharged. However, following the second cycle of chemotherapy, he was readmitted due to altered mental status. Due to financial constraints, the family decided not to pursue further treatment, leading to the patient’s eventual demise. Overall, this case highlights the critical importance of multidisciplinary collaboration for managing critically ill ANKL patients. Careful evaluation of the risks associated with chemotherapy, combined with timely administration of emergency chemotherapy in the ICU and comprehensive multi-organ support, can potentially offer a survival opportunity.
... However, in the 5th edition of the WHO Classification of Haematolymphoid Tumours, it is now recognized as a distinct entity of peripheral T-cell lymphoma, not otherwise specified [27]. This rare lymphoma frequently involves the skin and central nervous system and is associated with a grave prognosis [28]. Due to its unclear pathological nature, this subtype has been excluded from this analysis. ...
Article
Full-text available
Background Natural killer/T-cell lymphoma (NKTL) is a rare malignancy of mature natural killer/T-cells, predominantly found in Asian and South/Central American populations, with limited studies conducted in Europe and the United States. Objective The aim of this study is to present an overview of the incidence rate, demographic and clinical characteristics, treatment options, overall survival (OS), and factors influencing OS of NKTL in the United States. Methods We used data from the Surveillance, Epidemiology, and End Results 17 database to analyze NKTL cases recorded between 2000 and 2020. In a cohort of 1162 patients with NKTL, we calculated the incidence rates and performed statistical analyses to evaluate OS, the effect of radiotherapy and chemotherapy on survival, and lymphoma-specific survival. Results The mean annual incidence rate of NKTL in the United States was 0.067 per 100,000, with higher rates observed in men compared to women, and an increase noted with age. However, there has been no significant rise in incidence over recent years. Significant racial disparities were observed, with higher incidence rates in non-Hispanic Asian or Pacific Islanders and Hispanic people. The median survival time for patients with NKTL was 21 months, with a 5-year OS rate of 39.5%, which has shown improvement in recent years. Key independent prognostic factors impacting patient survival included age at diagnosis, clinical stage, nasal type presentation, presence of systemic symptoms, and treatment modality. Patients receiving combined radiotherapy and chemotherapy exhibited the best outcomes, with a median OS of 138 months and a 5-year OS rate of 58%. This survival benefit remained consistent even in patients with stage I/localized nasal type lymphoma, achieving a 5-year OS rate of 73.3%. Conclusions The incidence of NKTL has remained stable in recent years. Patients with the nasal type generally experience better survival outcomes. The use of combined radiotherapy and chemotherapy appears to enhance survival, though further validation through prospective multicenter clinical trials is necessary.
... Unfortunately, early attempts utilizing chemotherapeutic regimens of non-Hodgkin lymphomas have all ended with a dismal outcome in ANKL (5). The only potential cure is induction with L-asparaginase-based chemotherapy followed with allogeneic hematopoietic stem transplant (HSCT) (6). ...
Article
Full-text available
Background Aggressive natural killer (NK) cell leukemia (ANKL) is a rare NK cell neoplasm associated with Epstein-Barr virus (EBV) infection. Programmed cell death protein 1 (PD-1) blockade, which is successful in extranodal NK/T-cell lymphoma and EBV-related hemophagocytic lymphohistiocytosis, is considered to have a potential role in managing ANKL. Objectives This study aims to characterize ANKL clinically and evaluate the prognostic impact of anti-PD-1 antibody treatment. Methods We retrospectively analyzed the clinical characteristics and treatment regimens of ANKL patients from March 2009 to October 2023 in a single center. Data on clinical characteristics, treatment regimens and prognosis were collected from medical records. Overall survival (OS) of different risk groups was analyzed by Kaplan-Meier method. The least absolute shrinkage and selection operator (LASSO)-penalized Cox regression was used to identify the potential prognostic factors of ANKL. Results From March 2009 to October 2023 a total of 71 ANKL were retrieved with an OS of 2.0 months. Seven patients (9.9%) received PD-1 antibodies combined with various chemotherapies; thirty-five patients (49.3%) received asparaginase as part of chemotherapy; and eight patients (11.3%) received allogeneic HSCT after induction chemotherapy. Among patients who did not undergo allogeneic hematopoietic stem transplantation (HSCT), patients who received PD-1 antibodies as part of chemotherapy exhibited a superior OS than those without PD-1 antibodies (5.4 vs 1.6 months, p=0.035). The 1-year OS rate was 43% in the PD-1 subgroup compared with only 4% in the non-PD-1 subgroup. LASSO-Cox multivariate analysis revealed that PD-1 antibodies-containing regimens were associated with better survival (hazard ratio [HR]=0.349, 95% CI: 0.145~0.840, p=0.019). So was it with HSCT and asparaginase (HR=0.267, 95% CI=0.101~0.701 and HR=0.355, 95% CI=0.206~0.613, respectively). Conclusion ANKL still had a poor outcome in the past decade. Integration of anti-PD-1 antibody into chemotherapeutic therapy significantly improved the survival of ANKL. The prolonged survival attributed to PD-1 blockade could provide critical opportunities for patients awaiting HSCT.
... Targeted therapy with a monoclonal antibody against CD38 (Daratumumab) has been utilized with gleaming prospect, but still is not standard of care. [5,6] In the present case, daratumumab was utilized as a salvage therapy for progressive disease under the SMILE and CHOP regimen. e combination of daratumumab with the CHOP regimen resulted in excellent disease remission. ...
Article
Full-text available
Aggressive natural killer cell leukemia (ANKL) is a rare hematological malignancy, with only a few cases reported in the literature. The overall median survival is estimated to be less than 2 months. Moreover, Epstein–Barr virus (EBV)-negative ANKL is a rarity, with only a few being EBV-negative. We present a 27-year-old male with pancytopenia, massive splenomegaly, and no lymphadenopathy. Bone marrow (BM) aspirate flow cytometry revealed abnormal lymphocytes showing moderate expression of CD56, CD16, CD38, CD2, and CD45, dim expression of CD7 and CD117, and variable cytoplasmic CD3 and were negative for immaturity markers and other T lymphocytic, myeloid, and plasmacytoid dendritic cell markers. The BM biopsy revealed negative terminal deoxynucleotidyl transferase, confirming the diagnosis of ANKL. EBV was negative; hence, the final diagnosis of EBV-negative ANKL was offered. The patient had a progressive course despite two cycles of modified SMILE regimen and CHOP. The addition of daratumumab (anti-CD38 monoclonal antibody) to CHOP resulted in disease remission. The patient has survived for more than 18 months after diagnosis and is presently on follow-up on maintenance daratumumab, awaiting an allogenic stem cell transplant. This article aims to report rare refractory EBV-negative ANKL patients successfully managed with an unconventional regimen; hence, further studies are warranted for targeted therapy in fulminant diseases like ANKL.
... For ANKL, management typically involves chemotherapy and stem cell transplantation. Promising therapeutic agents include BCL-2 inhibitors, immune checkpoint inhibitors like PD-1/PD-L1 antibodies, JAK1 and JAK2 inhibitors, and histone deacetylase inhibitors [8,28]. The patient of the present case was planned for stem cell transplant but unfortunately developed multi-organ failure and passed. ...
Article
Full-text available
A 24-year-old Ecuadorian female, previously diagnosed with acute fatty liver (AFL) during pregnancy, developed constitutional symptoms, jaundice, and abdominal pain in a subsequent pregnancy, prompting investigations that suggested a recurrence of AFL. She underwent an elective abortion, which resulted in the resolution of her abdominal pain, and a liver biopsy, which showed granulomatous inflammation and lymphocytic infiltration. She later presented with abdominal distention, productive cough, and persistent constitutional symptoms and jaundice. Extensive laboratory and imaging studies indicated sepsis, acute liver injury, and disseminated intravascular coagulopathy. Her serum Epstein-Barr virus (EBV) level was elevated. Special staining of her previous liver biopsy revealed EBV-positive natural killer (NK) cells. A bone marrow biopsy also revealed EBV-positive NK cells. She was diagnosed with aggressive NK cell leukemia (ANKL) with or without chronic active EBV (CAEBV). Treatment included dexamethasone, atovaquone, bortezomib, and ganciclovir, with plans for a stem cell transplant. However, her course was complicated by infections and multi-organ failure, resulting in her passing. This case highlights the rarity and challenges in managing EBV-associated ANKL, emphasizing the need for early detection and improved treatment options, with stem cell transplantation offering the best prognosis.
Chapter
Master the implementation of flow cytometry techniques in diagnosing complex hematologic diseases and malignancies in patients with this comprehensive and practical guide. Featuring updated recommendations on pre-analytical steps, instrument settings and panel construction, this manual offers invaluable support for those diagnosing, treating and researching hematologic malignancies. Written by leading experts in the field, this book puts flow cytometry into everyday context. Through engaging with differential diagnoses, the manual offers an accurate description of specific features of malignancies and mimicking conditions. It also places flow cytometry in the scope of integrated diagnosis according to WHO and ICC classifications. Fully updated throughout, this second edition provides new literature and concepts, and presents new AI-based analytical methods. Richly illustrated and in full colour, this book is an ideal resource for those working in flow cytometry laboratories.
Article
Lymphoproliferative disorders of natural killer (NK)-cell lineage are well documented in humans but have yet to be documented in non-human primates (NHPs). Here we describe a case of NK-cell lymphoproliferative disorder/leukemia in a 20-y-old captive female rhesus macaque ( Macaca mulatta). The animal clinically had mild splenomegaly and marked lymphocytosis with small-to-medium lymphocytes in blood smears. By flow cytometry and cluster differentiation, the lymphocytes were CD3-negative, CD8-positive, CD4-negative, and CD20-negative for cell surface markers; immunohistochemistry revealed the presence of intracellular CD3 and granzyme B. This immunoprofile is consistent with a NK-cell phenotype. Histologically, these cells were predominantly intravascular within the splenic red pulp, liver sinusoids, and to a lesser degree bone marrow. Oncogenic viruses, such as Mason-Pfizer monkey viruses (MPMV; formerly, and commonly known as, simian retroviruses or SRV; Retroviridae, Betaretrovirus maspfimon); simian immunodeficiency virus (SIV; Retroviridae, Lentivirus simimdef), and primate T-lymphotropic virus 1 (PTLV1; commonly known as simian T-lymphotropic virus type 1, STLV1; Retroviridae, Deltaretrovirus priTlym1), were not detected in this animal by serology. Immunohistochemistry using EBNA2 antibody to detect rhesus and cynomolgus monkey lymphocryptovirus (McGHV4/RLV and McGHV10 respectively; Orthoherpesviridae, Lymphocryptovirus macacinegamma4 and Lymphocryptovirus macacinegamma13, respectively) was negative. Together these findings are consistent with a diagnosis of naturally occurring NK-cell lymphoproliferative disorder. NK-cell lymphoproliferative disorder has not been reported previously in rhesus macaques, to our knowledge.
Article
Full-text available
Cutaneous T-cell lymphoma (CTCL) is a form of non-Hodgkin lymphoma that manifests initially in the skin and disseminates systemically as disease progresses. Mycosis fungoides and Sézary syndrome (MF/SS) are the most common subtypes of CTCL. Advanced MF/SS are life-threatening with few treatment options. We searched for new agents by high-throughput screening of selected targeted compounds and identified high value targets including phosphatidylinositol 3-kinase (PI3K) and cyclin-dependent kinases. To validate these hits from the screen, we developed patient-derived xenograft (PDX) mouse models that recapitulated the cardinal features of MF/SS and maintained histologic and molecular characteristics of their clinical counterparts. Importantly, we established a blood-based biomarker assay using tumor cell-free DNA to measure systemic tumor burden longitudinally in living mice during drug therapy. A PI3K inhibitor, BKM120, was tested in our PDX model leading to disease attenuation and prolonged survival. Isoform-specific siRNA knockdowns and isoform-selective PI3K inhibitors identified PI3K-δ as required for tumor proliferation. Additional studies showed synergistic combination of PI3K-α/δ inhibitors with histone deacetylase (HDAC) inhibitors. The strong preclinical efficacy of this potent combination against multiple PDX models makes it an excellent candidate for further clinical development.
Article
Full-text available
Aggressive NK cell leukemia (ANKL) is a rare malignant lymphoproliferative disorder of mature NK cells closely associated with Epstein-Barr virus (EBV) and more common in East Asia than in other areas. Significant variations exist in the morphology of ANKL tumor cells, from typical large granular lymphocyte morphology to highly atypical features with basophilic cytoplasm containing azurophilc granules. The main involved sites are hepatosplenic lesions, bone marrow and peripheral blood, and nasal or skin lesions are infrequent. A fever and liver dysfunction with an often rapidly progressive course are the main clinical symptoms, including hemophagocytic syndrome and disseminated intravascular coagulation. Although the outcome had been dismal for decades, with a median survival of less than three months, the introduction of combined chemotherapy including L-asparaginase and allogeneic hematopoietic cell transplantation has helped achieve a complete response and potential cure for some patients. With the advent of next-generation sequencing technologies, molecular alterations of ANKL have been elucidated, and dysfunctions in several signaling pathways, including the JAK/STAT pathway, have been identified. Novel target approaches to managing these abnormalities might help improve the prognosis of patients with ANKL.
Article
Full-text available
Aggressive natural killer-cell (NK-cell) leukemia (ANKL) is an extremely aggressive malignancy with dismal prognosis and lack of targeted therapies. Here, we elucidate the molecular pathogenesis of ANKL using a combination of genomic and drug sensitivity profiling. We study 14 ANKL patients using whole-exome sequencing (WES) and identify mutations in STAT3 (21%) and RAS-MAPK pathway genes (21%) as well as in DDX3X (29%) and epigenetic modifiers (50%). Additional alterations include JAK-STAT copy gains and tyrosine phosphatase mutations, which we show recurrent also in extranodal NK/T-cell lymphoma, nasal type (NKTCL) through integration of public genomic data. Drug sensitivity profiling further demonstrates the role of the JAK-STAT pathway in the pathogenesis of NK-cell malignancies, identifying NK cells to be highly sensitive to JAK and BCL2 inhibition compared to other hematopoietic cell lineages. Our results provide insight into ANKL genetics and a framework for application of targeted therapies in NK-cell malignancies.
Article
Aggressive natural killer-cell leukemia (ANKL) is a rare, lethal disease with pathologic features that are underdescribed in the literature, particularly in Western nations. In addition, although data on the molecular pathogenesis of ANKL has been reported, evaluation of such data in a clinicopathologic context remains limited. Patients diagnosed with ANKL were identified retrospectively. Detailed demographic and clinicopathologic data were analyzed. We assessed novel markers by immunohistochemistry and performed targeted next-generation sequencing analysis. The study group included 9 men and 3 women with a median age at diagnosis of 47.5 years (range, 20 to 75 y). Two distinct patterns of bone marrow involvement were identified: interstitial and sinusoidal. The neoplastic cells were positive for CD56 and CD94, and negative for surface CD3, CD5, and CD57 in all cases assessed. They were also positive for CD2 (10/12), c-MYC (6/8), BCL2 (6/8), CD16 (5/7), EBER (9/12), CD7 (6/11), pSTAT3 (3/8), CD8 (2/6), PD-L1 (2/8), CD4 (2/11), CD8 (2/6), and CD158 (1/5). Aberrant p53 expression was identified in most (7/8) cases; p53 was strongly expressed in 4 cases. Conventional cytogenetic analysis showed clonal abnormalities in 5 of 12 cases. TP53 mutations were detected in 3 of 6 cases, whereas ASXL1 and TET2 mutations were each detected in 2 of 6 cases. Patients had very poor outcomes despite intensive chemotherapy, with a median survival of 2 months. ANKL exhibits 2 distinct patterns of tissue involvement. Neoplastic cells in ANKL are commonly positive for c-MYC and EBER, and they have a high frequency of p53 overexpression, frequently with corresponding TP53 mutations.
Article
A 14-year-old Japanese female with neutropenia showed malignant proliferation of the large granular lymphocytes (LGLs). These LGLs were E rosette+ and Fc(IgG) receptor+ and therefore are referred to as T gamma lymphocytes. They were also Leu-11+ and OKT11+; however, they were clearly negative for Leu-7, OKT3, OKT8, OKM1, and HNK-1 antigens as well as for terminal deoxynucleotidyl transferase activity. Karyotype analysis revealed 47, XXX. The LGLs showed no rearrangement of T cell receptor C beta genes. The natural killer (NK) cell activity against K562 target cells was low, but was significantly augmented after stimulation by recombinant human interleukin 2 (IL 2) in contrast to minimal NK boosting by recombinant human gamma-interferon (gamma- IFN). Such a unique responsive ability to lymphokines was quite similar to that noted in fetal and cord blood cells. These LGLs also demonstrated a considerable increase in antibody-dependent cell- mediated cytotoxicity (ADCC) and lymphokine-activated killer (LAK) activity after a short incubation with IL 2. Although in a resting stage they showed no IL 2 receptor expression as examined by anti-Tac antibody, Tac antigen appeared after IL 2 treatment followed by a marked increase in 3H-thymidine incorporation and a remarkable production of gamma-IFN. To investigate the mechanism of neutropenia, in vitro IL 2-stimulated coculture studies of these cells with normal bone marrow cells were performed. Colony formation of myeloid progenitors (CFU-C) was significantly suppressed. In addition, the conditioned medium from IL 2-stimulated LGLs indicated a remarkable suppression of CFU-C. These results suggest that these LGLs with a Leu- 11+, Leu-7- surface phenotype might belong to a unique subset of pre-NK cells that are functionally and phenotypically similar to those represented at any early stage of human ontogeny and that they strongly express Tac antigen under the influence of IL 2 administration, followed by remarkable cell proliferation and gamma-IFN production.
Article
There have been many reports of cases in which chronic increases in the numbers of natural killer (NK) cells have been reported. Whether this is reactive or neoplastic in nature has been debated. We report the first case of an aggressive NK cell leukemia in an adult with establishment of an NK cell line. A 70-year-old man had two spontaneous episodes of jejunal perforation and one month later developed a severe febrile illness with moderate splenomegaly. Hemoglobin was 13.1 g/L, and WBC count was 1.8 X 10(9)/L with 2% large granular lymphocytes (LGLs). Platelet count was 143 X 10(9)/L; prothrombin time (PT) and partial thromboplastin time (PTT) were normal. Bone marrow was infiltrated with 25% to 30% LGLs; serum lysozyme was normal. Serum LDH was initially 1,191 U/L and rose to 6,408 (normal 240 to 525 U/L). Ten days later, the WBC count increased to 99.9 X 10(9)/L with 70% LGL cells; the PT and PTT increased, and the platelet count dropped. No bacterial or viral cause of fever was identified. The cells from peripheral blood were LGLs that stained positively for acid phosphatase. All of the LGLs reacted with a monoclonal antibody reactive with NK cells (LEU-11b). Functionally, the patient's peripheral blood mononuclear cells (PBMs) demonstrated 100 times more lytic activity against K562 tumor cell lines than did normal PBMs. The patient's PBMs were propagated in vitro. The cultured cells showed the morphological, cytochemical, immunological, and functional characteristics of NK cells. In addition, partial trisomy involving chromosome 1 q with duplication in regions of q21 through q31 was observed in all metaphases analyzed. The extra chromosome 1q with duplication in regions q21 through q31 was translocated to the p- terminal of chromosome 5. One percent to 5% of normal PBMs comprise NK cells; in most cases, leukemias arise from normal phenotypic counterparts. This case demonstrated that aggressive NK cell leukemia may occur in adults. In addition, the chromosomal abnormalities suggest that this is not a reactive process but a malignancy.
Article
Background: Chronic lymphoproliferative disorder of NK-cells (CLPD-NK) manifests as a persistent increase (≥2 x 109 /L, for > 6 months) of mature NK-cells in peripheral blood with an indolent clinical course. The disease is rare, and only limited case series have been published. Methods: We retrospectively studied 11 patients with CLPD-NK diagnosed at our institution between 2005 and 2017. Results: Patients included 7 men and 4 women (median age, 60 years). Ten patients (91%) had cytopenias. Bone marrow involvement was limited (5-15%). The most commonly detected antigenic aberrancies by flow cytometry immunophenotyping were: CD7 decreased/dim (30%), CD8uniform + (36%), CD56-/partial (73%), CD94bright (55%), and KIR restriction (100%). JAK/STAT pathway mutations were detected in 8/10 (80%) patients, STAT3 (n=7) and JAK3 (n=1). The mutations tended to correlate with more frequent and greater severity of anemia and thrombocytopenia, and treatment requirements. Seven patients received single-agent therapy, with amelioration of symptoms; 4 patients were observed. There were no disease-associated deaths or progression to more aggressive disease during the follow up interval (median, 17 months). Conclusions: Patients with CLPD-NK have an indolent clinical course and frequent hematologic manifestations that are responsive to single-agent therapy. Mutations in STAT3 are common and portend more pronounced clinical manifestations. This article is protected by copyright. All rights reserved.
Article
Aims: Distinguishing between aggressive NK-cell leukaemia (ANKCL) and extranodal NK/T-cell lymphoma (ENKTCL) with secondary bone marrow involvement is challenging, as they are rare bone marrow NK/T-cell neoplasms and share similar features. Methods and results: We studied bone marrow NK/T-cell neoplasms by classifying them into those with no extramedullary mass (group 1, 8 cases) and those with extramedullary mass (group 2, 13 cases). Both groups showed similar clinical presentations and pathological features. Fever and cytopenia were the most common clinical presentations in both groups. The neoplastic cells varied from small and relatively monotonous cells to large pleomorphic cells. In six cases (2 in group 1 and 4 in group 2), the neoplastic infiltrate was inconspicuous, consisting of ≤10% of marrow cells in the interstitium, which were hardly identified by H&E stain alone. Nearly all patients were rapidly fatal no matter the neoplastic infiltrate volume. All the patients in group 1 fulfilled the WHO 2017 diagnostic criteria of ANKCL and their survivals were significantly worse than those of the group 2 patients (P = 0.035). In addition, group 1 patients were significantly associated with chromosome 7 abnormalities. Chromosome 6q deletion, commonly reported in ENKTCL, was seen in two of our group 2 patients, and was not observed in any of our group 1 patients. Conclusion: ANKCL with no extramedullary mass should be distinguished from ENKTCL with bone marrow involvement as the former showed distinct outcome and genetic features. This article is protected by copyright. All rights reserved.