You are on page 1of 9

Leukemia (2011) 25, 101–109

& 2011 Macmillan Publishers Limited All rights reserved 0887-6924/11


www.nature.com/leu

ORIGINAL ARTICLE

Analysis of CD16 þ CD56dim NK cells from CLL patients: evidence supporting a


therapeutic strategy with optimized anti-CD20 monoclonal antibodies
M Le Garff-Tavernier1,2,3, J Decocq1,3,4, C de Romeuf4, C Parizot5, CA Dutertre4,6,7,8, E Chapiro2, F Davi1,2,3, P Debré1,3,5,
JF Prost4, JL Teillaud6,7,8, H Merle-Beral2,6,7,9 and V Vieillard1,3,9
1
INSERM, UMR-S 945, Paris, France; 2AP-HP, Hôpital Pitié-Salpêtrière, Service d’Hématologie Biologique, Paris, France;
3
UPMC Université Paris 06, UMR-S 945, Paris, France; 4Laboratoire Français de Fractionnement et des Biotechnologies (LFB),
Les Ulis, France; 5Laboratoire d’Immunologie Cellulaire et Tissulaire, AP-HP, Hôpital Pitié-Salpêtrière, Paris, France;
6
INSERM, UMR-S 872, Paris, France; 7Centre de Recherche des Cordeliers, UPMC Université Paris 06, UMR-S 872,
Paris, France and 8Université Paris Descartes, UMR-S 872, Paris, France

Although anti-CD20 monoclonal antibodies (mAbs) show ‘gold standard’ for the treatment of CLL,1,2 it does not allow
promise for the treatment of chronic lymphocytic leukemia full-disease eradication and the disease can become resistant.
(CLL), the success of the anti-CD20 mAb rituximab in CLL The activity of rituximab is thought to be mediated through
treatment has been limited. Novel anti-CD20 mAbs with more
potent cytotoxic activity have recently been engineered, but so several mechanisms, including complement-dependent cyto-
far most have only been tested in vitro with natural killer (NK) toxicity, apoptosis, antibody-dependent cellular cytotoxicity
cells from healthy donors. Because it is still unclear whether (ADCC) and/or phagocytosis.3 Strategies pursued to improve
these optimized cytotoxic mAbs will improve NK-cell killing of the efficacy of mAb therapy have included modifying the
tumor cells in CLL patients, we characterized the relevant antibody structure to increase its affinity to the Fc gamma
phenotypic and functional features of NK cells from CLL receptor IIIA (FcgRIIIA, CD16), an intermediate-affinity activat-
patients in detail. Expression of inhibitory and activating
NK-cell receptors and of Fc gamma receptor IIIA (FccRIIIA) is ing FcgR strongly expressed on natural killer (NK) cells.4
well preserved in CD16 þ CD56dim cytotoxic NK cells from these Optimized mAbs, including AME-133, GA-101, veltuzumab
patients, independently of disease progression. These cells are (hA20) and ofatumumab (HuMax-CD20)5–7 are at different
fully functional following cytokine stimulation. In addition, stages of development. We previously described a novel
the FccRIIIA-optimized LFB-R603 anti-CD20 mAb mediates 100 chimeric anti-CD20 mAb, EMAB-6, characterized by low fucose
times greater antibody-dependent cell-mediated cytotoxicity by content in its Fc region. EMAB-6 shows improved FcgRIIIA
NK cells from CLL patients and healthy donors than rituximab.
Enhanced degranulation against autologous B-CLL cells is binding, resulting in enhanced ADCC, mediated particularly by
observed at lower concentrations of LFB-R603 than rituximab, macrophages8 and NK cells.9
regardless of CLL prognostic factors. These findings strongly The NK cells constitute a unique component of the innate
justify further clinical development of anti-CD20 mAbs immune system, able to recognize various targets without
optimized for FccR engagement in CLL patients. specific sensitization. The NK cells are heterogeneous and
Leukemia (2011) 25, 101–109; doi:10.1038/leu.2010.240; differ in their proliferative potential, homing characteristics,
published online 26 October 2010
Keywords: chronic lymphocytic leukemia (CLL); natural killer (NK) functional capabilities and in responses to a wide range of
cells; anti-CD20; monoclonal antibodies cytokines. Around 10% of NK cells in the peripheral blood are
CD16CD56bright. This immunoregulatory subset of NK cells
produces a wide range of cytokines and chemokines, but its
ability to kill target cells spontaneously is poor. In contrast, the
rest (90%), corresponding to the CD16 þ CD56dim cytotoxic
NK-cell subset, produces relatively lower cytokine levels, but
Introduction possesses an abundance of cytolytic granules and can sponta-
neously lyse susceptible target cells.10 The complexity of the
Chronic lymphocytic leukemia (CLL) is an accumulation NK-cell compartment is attributable to the vast network
of CD5 þ /CD19 þ malignant monoclonal B lymphocytes with of inhibitory or activating receptors that allow these cells to
a highly variable clinical course. The introduction of therapeutic recognize target cells. The functional state of NK cells is,
monoclonal antibodies (mAbs) has brought a remarkable change therefore, dictated by the balance between opposing signals.11
in treatment. The anti-CD52 alemtuzumab and anti-CD20 Moreover, in the presence of mAbs, strong expression of
rituximab mAbs have been widely used in progressive disease. FcgRIIIA allows CD16 þ CD56dim NK cells to kill targets through
In contrast to alemtuzumab, rituximab is generally well tole- ADCC, regardless of the presence of any inhibitory receptors.
rated with infrequent serious adverse effects that are generally ADCC has been identified as a critical mechanism underlying
well controlled. Although the combination of fludarabine and the clinical efficacy of therapeutic antibodies, including
cyclophosphamide with rituximab is currently considered the rituximab.12,13 We focused our attention on NK cells in patients
with CLL, because the size of this cell compartment is an
Correspondence: Dr M Le Garff-Tavernier, INSERM UMR-S 945, independent factor predictive of the rate of disease progression
Bâtiment CERVI, GH Pitié-Salpêtrière, 83 bd de l’Hôpital, 75651 Paris in patients with newly diagnosed CLL.14 Several studies have
Cedex 13, France. reported functional alterations in NK cells in these patients,
E-mail: magali.legarff@psl.aphp.fr
9
These authors are senior co-authors. including defective cytotoxic activity, which are particularly
Received 19 February 2010; revised 12 August 2010; accepted 1 pronounced in advanced disease and that could be because of
September 2010; published online 26 October 2010 the accumulation of leukemic cells rather than to any intrinsic
NK cells and optimized anti-CD20 in CLL
M Le Garff-Tavernier et al
102
defect in the former.15,16 This hypothesis was suggested by the CD335/NKp46 (BAB281), CD85j/ILT2 (HP-F1), 2B4 (C1.7) and
restoration of their functional activity after interferon (IFN)-a CD69 (TP1.55.3) (Beckman-Coulter), CD16 (3G8) and LAIR1
and/or interleukin (IL)-2 treatment.17,18 (DX26) (BD Biosciences), NKG2C (134591) and NKp80/KLFR1
We conducted the first extensive characterization of the (239127) (R&D Systems, Lille, France) and CD337/NKp30
phenotypic and cytotoxic qualities of NK cells from CLL patients (AF29-4D12) (Miltenyi Biotec, Paris, France). Erythrocytes were
and compared them with those of healthy subjects. Our data lysed with a FACS lysing solution kit (BD Biosciences). After
reveal that NK cells from these patients have the major extensive washing, at least 5  103 CD3CD56dim NK cells
phenotypic features of competent NK cells and are able were analyzed on a FACSCanto I with FACSDiva 5.03 software
to function effectively. We also explored the preclinical activity (BD Biosciences). The level of CD16 expression on
of LFB-R603, a novel optimized anti-CD20 mAb, and show that CD3CD56dim NK cells was quantified using QuantiBRITE
at low doses this antibody promotes a significantly higher standard beads (BD Biosciences), as recommended by the
percentage of NK cell-mediated degranulation against auto- manufacturer. For intracellular IFN-g staining, peripheral blood
logous CLL cells than rituximab. mononuclear cells (PBMC) were incubated overnight in the
presence of IL-12 (10 ng/ml) and IL-18 (100 ng/ml) (R&D
Systems). Cells were fixed and permeabilized with a cytofix/
Patients and methods cytoperm kit (BD Biosciences), and stained with IFN-g mAb
(B27; BD Biosciences), as described.21
Patients and control donors
Fresh blood samples were collected from 57 CLL patients
followed at Pitié-Salpêtrière Hospital (Paris, France). All patients Direct cytolytic activity of NK cells
met the morphological and phenotypic criteria for typical NK cell-enriched PBMC (8 CLL patients and 5 controls) were tested
CLL (Matutes score X4).19 They had not been treated previously for direct cytotoxicity. NK cell-enriched PBMC fractions were
or had not received any treatment during the 6 months before obtained after negative depletion (anti-CD19-coated beads)
the study. None of these patients had received rituximab or followed by positive selection (anti-CD56-coated beads) (Miltenyi
alemtuzumab. Binet stage A patients were considered to have an Biotec, Bergish Gladbach, Germany). Similar proportions of NK
unfavorable prognosis if they met any two of the following four cells were detected in NK cell-enriched PBMC fractions from CLL
criteria: (1) Presence of unmutated Immunoglobulin heavy patients (38±11%) and controls (42±10%). The B cells accounted
variable chain sequences with a VH gene sequence p2% of for 30±20% of the residual cells for CLL patients and 1±2% for
sequence alterations compared with the published germline controls, whereas the proportions for T cells were 31±17% and
sequence; (2) High expression of surface CD38 (420%); (3) 55±11%, respectively. Direct cytotoxicity was assessed after IL-2
High expression of cytoplasmic ZAP-70 (420%); and (4) Clonal stimulation (proleukin-2, 1000 IU/ml) for 72 h, with a standard 4-h
51
cytogenetic abnormalities, including deletion of chromosomes Cr-release assay against the human leukocyte antigen (HLA) class
17p or 11q or trisomy of chromosome 12, revealed by I-deficient human K562 cell line.21
fluorescent-labeled DNA probes used in interphase fluorescence
in situ hybridization (probes 17p13.1, ATM 11q23 and CEP12,
Anti-CD20 LFB-R603 antibody
respectively).20 Patients with del (13q) (probe 13q14), have an
LFB-R603 is a chimeric mAb directed against CD20 with a
excellent prognosis if they do not have additional defects.
glycosylation profile that allows increased binding to CD16.
Patients with 17p were considered to have an unfavorable
This mAb has been granted an orphan-drug status in Europe and
prognosis even if all their other criteria were favorable. All other
in the USA for CLL treatment (http://www.lfb.fr/en/home.html).
Binet stage A patients were considered to have a favorable
A previous generation of this antibody, named EMAB-6, sharing
prognosis. The control group comprised sex- and age-matched
the same specificity and a similar glycosylation pattern, was
outpatient volunteers from the rheumatology (n ¼ 29), who
described in de Romeuf et al.9
had consulted for noninflammatory arthritic pathology such
as osteoarthritis, or gerontology (n ¼ 38) departments at Pitié-
Salpêtrière or Charles-Foix Hospital, respectively. Our study Purification of NK cells and ADCC assay
applied restrictive health selection criteria, including no Purified NK cells (498% CD3CD56 þ cells) from 7 CLL
infectious or malignant diseases 6 months before the study patients and 6 controls were obtained from peripheral blood
and without acute illness at the time of sampling. None of the after centrifugation on a Ficoll/Hypaque gradient, followed by
control patients had an autoimmune disease. All case and negative selection with anti-CD19-coated beads (Miltenyi
control patients provided a written, informed consent before Biotec) and cytofluorometric selection of CD3CD56 þ NK
participation. The local ethics committee approved the study. cells on a FACSVantage (BD Biosciences). ADCC assays were
performed with purified NK cells and 51Cr-labeled Raji cells at
an effector-target (E:T) ratio of 10:1. Experiments were
Flow cytometry and absolute B cell, T cell, and performed in the presence of LFB-R603 (LFB) or rituximab
NK cell counts (Roche Ltd, Basel, Switzerland) at concentrations ranging from
Freshly collected blood cells underwent cell-surface staining for 0.1 to 10 000 ng/ml.
four-color cytometric analysis. Isotype-matched immunoglobulin
served as the negative control. NK cells were analyzed after
staining with an appropriate antibody cocktail: CD3-FITC Degranulation of NK cells
(UCHT1; Beckman-Coulter, Marseille, France), CD19-PerCP The degranulation marker CD107a was detected on freshly
Cy5.5 (SJ25C1; BD Biosciences, Le Pont de Claix, France), isolated PBMC from both CLL patients and controls by
CD56-APC (N901; Beckman-Coulter), and the following PE- co-staining for NK markers (CD3CD56 þ NK cells). Cells were
conjugated antibodies: KIR2DL1/KIR2DS1 (EB6B), KIR2DL2/ incubated with anti-CD107a mAb (H4A3; BD Biosciences),
KIR2DL3/KIR2DS2 (GL183), KIR3DL1/KIR3DS1 (Z27), CD159a/ with 10 or 1000 ng/ml of either anti-CD20 rituximab (Roche) or
NKG2A (Z199), NKG2D (ON72), CD336/NKp44 (Z231), LFB-R603 (LFB, Les Ulis, France), as described.21

Leukemia
NK cells and optimized anti-CD20 in CLL
M Le Garff-Tavernier et al
103
Statistical analysis Phenotypic analysis of NK receptors in CLL patients
Statistical analyses were performed with Prism 4 software The phenotypic properties of the CD3CD56dim cytotoxic NK-
(GraphPad Software, San Diego, CA, USA). Intergroup cell subset were investigated further by gating out CD3CD56bright
comparisons were assessed with the Mann–Whitney U-test or NK cells. CD56dim NK cells from CLL patients, irrespective of their
Wilcoxon rank-sum test. P-values less than 0.05 were considered prognostic status, were indistinguishable from those of controls in
statistically significant; *Po0.05, **Po0.01, ***Po0.001. terms of cell-surface expression of the major NK receptors studied,
including c-lectin receptors (NKG2A, NKG2C and NKG2D),
natural cytotoxicity receptors (NKp30, NKp44 and NKp46) and
Results other NK-cell markers such as NKp80, ILT-2, LAIR-1 and 2B4
(Figure 2a, upper panels; data not shown). Similar phenotypic
CLL patients results were observed according to the Binet stages (A vs B/C) (data
Table 1 and Supplementary Table 1 show the main clinical and not shown). This phenotype was then studied longitudinally, over a
biological prognostic characteristics of CLL patients included in 1-year period, comparing the initial samples with samples taken 2,
this study. Because the potential benefit of early treatment for 4, 6 and/or 12 months later. A remarkable phenotypic stability was
unfavorable prognosis Binet stage A patients is a key therapeutic observed for each NK receptor studied. Figure 2a (lower panels)
question, patients were divided into two groups according to shows representative CLL patients from each of the groups (patients
their biological prognostic factors. Binet stage A CLL patients 1 and 2, group 1; patient 3, group 2; patient 4, group 1 (at first
with an unfavorable prognosis were classified with Binet stage evaluation) and group 2 (at 12 months)). These results were
B/C patients (group 2, n ¼ 23) and compared with stage A confirmed with three other patients (data not shown). In addition,
patients with a favorable prognosis (group 1, n ¼ 34). There was patient 4’s NK receptor repertoire remained stable during disease
no significant difference in the sex ratio or median age between progression between stages A (group 1) and C (group 2) after 1
groups 1 and 2. year. The only difference observed in the KIR was a weak, but
significant decrease in expression of KIR2DL1/DS1, which
recognizes group-2 HLA-Cw alleles, in CLL patients with an
unfavorable prognosis compared with controls (P ¼ 0.0370). Binet
Distribution of lymphocyte subsets in CLL patients B/C patients presented a nonsignificant decrease compared with
Flow cytometry was used to measure CD19 þ B, CD3 þ T and controls (data not shown). No differences between groups were
CD3CD56 þ NK subsets in CLL patients and controls. The observed for the other KIRs tested, including KIR2DL2-3/DS2,
B-cell counts were significantly higher in CLL patients (group 1: which binds group-1 HLA Cw ligands, and KIR3DL1/DS1, which
median 10 592/mm3 (range: 317–103 402); group 2: 28 084/ recognizes HLA-Bw4 (Figure 2b).
mm3 (2918–198 407)) than in controls (median 111/mm3 The expression pattern of the early activation marker CD69 on
(22–943)) (groups 1 and 2 vs controls: Po0.0001). The CD3CD56 þ NK cells was also investigated. As shown in
distribution of lymphocyte subsets showed that CLL patients Figure 2c, CD69 expression in CD3CD56dim NK cells was
had significantly lower T-cell and NK-cell frequencies than slightly, but significantly lower in CLL patients than in controls
controls (groups 1 and 2 vs controls: Po0.0001 for T cells and (group 1 vs controls: P ¼ 0.0228, and group 2 vs controls:
Po0.0001 for NK cells) (Figures 1a and c, respectively). P ¼ 0.0044), even if a certain degree of overlap of CD69
Interestingly, CLL patients had significantly higher absolute expression in CD3CD56dim NK cells was observed between
T-cell and NK-cell counts than controls (groups 1 and 2 vs both groups of CLL patients and controls. Similarly, CD69
controls: Po0.0001 for T cells, group 1 vs controls: P ¼ 0.0066 expression decreased significantly in CD3CD56bright immuno-
and group 2 vs controls: P ¼ 0.0003 for NK cells) (Figures 1b and regulatory NK cells from group 1 patients compared with controls
d, respectively). However, despite higher lymphocyte counts in (P ¼ 0.0037). Furthermore, HLA-DR expression, a late cell-activa-
group 2 patients, T-cell and NK-cell counts did not differ tion marker, remained close to background levels in all control and
significantly between the two groups of CLL patients. Similarly, CLL subjects, in all NK-cell subsets (data not shown). Similar results
the distribution of the CD56dim NK-cell subset did not differ were obtained according to Binet stages (data not shown).
between the CLL groups and controls (Figure 1e). Finally, there
was no difference in the frequency of CD16 þ /CD56dim NK cells
(Figure 1f) or surface density of CD16, as detected by the Functional analysis of NK cells in CLL patients
Quantibrite assay (data not shown), between these groups. The functional capacities of the NK-cell compartment of CLL
Similar results were obtained using the classical stage A vs patients were investigated further. As shown in Figure 3a,
stages B/C of the Binet classification (data not shown). intracellular IFN-g in CD3CD56dim NK cells from CLL patients

Table 1 Main clinical and biological characteristics of CLL patients

Patients Age median Lymphocytosis Prognostic factors


(n) (range) (109/l) median
(range)
Sex Binet IgHV CD38 ZAP-70 Cytogenetic
stage mutational abnormalities
status

F M A B/C Ma UMa  +  + 17p  11q  13q  12 +

Group 1 (34) 11 23 62 (34–83) 14.9 (5.1–106.6) 34 0 31/31 0/31 27/34 7/34 27/31 4/31 0/33 0/33 18/33 1/33
Group 2 (23) 8 15 66 (40–79) 33.8 (7.3–212.2) 9 14 7/21 14/21 15/23 8/23 11/22 11/22 4/23 6/23 12/23 2/23
Abbreviations: CLL, chronic lymphocytic leukemia; F, female; IgHV, immunoglobulin variable heavy chain; M, male.
a
M, mutated; UM, unmutated.

Leukemia
NK cells and optimized anti-CD20 in CLL
M Le Garff-Tavernier et al
104
*** ***
*** 4 ***
100 10

75

CD3+/mm3
% CD3+
103
50

25
102
0 1
Ctl group 1 group 2 Ctl group 1 group 2

*** ***
*** **
40 4
* 10

CD3-CD56+/mm3
% CD3-CD56+

30
103

20
102
10
101
0 1
Ctl group 1 group 2 Ctl group 1 group 2

100 100
% CD56dim/CD3-CD56+

% CD16+/CD3-CD56dim

75 75

50 50

25 25

0 0
Ctl group 1 group 2 Ctl group 1 group 2
Figure 1 Distribution of lymphocyte subsets in CLL patients. FACS analysis of percentages and absolute cell counts of: (a and b) CD3 þ T cells;
and (c and d) CD3CD56 þ NK cells, gated on lymphocytes from peripheral blood. (e) Expression of CD56dim gated on CD3CD56 þ NK cells.
(f) Expression of CD16 þ gated on CD3CD56dim NK cells. Cells were collected from controls (Ctl, J) and CLL patients in group 1 (}) and group 2
(&). Horizontal bars represent the median values. P-values refer to the comparison between controls and CLL patients, assessed with the
Mann–Whitney U-test. *Po0.05, **Po0.01, ***Po0.001.

(groups 1 and 2) treated with IL-12 and IL-18 was expressed at a maximum ADCC was detected at 1 ng/ml of LFB-R603, but at
similar level to that in NK cells from controls. Similar results 100 ng/ml of rituximab. This indicates that the potency of
were observed in the CD3CD56bright NK-cell subset even LFB-R603 is superior to that of rituximab (Figure 4a). At 1 ng/ml,
if IFN-g expression increased slightly in CLL patients (group 1 the median ADCC value with LFB-R603 was 62.5% for controls
and group 2) compared with controls (P ¼ 0.0359 and and 65.1% for CLL patients, whereas at a similar dose of
P ¼ 0.0176, respectively) (Figure 3b). The direct cytotoxicity of rituximab, the ADCC was significantly lower (P ¼ 0.03) at 7.3
NK cells was then evaluated after IL-2 activation by their and 8.5%, respectively (Figure 4b, upper panel). In contrast, the
capacity to lyse HLA class-I negative K562 sensitive-target cells ADCC with both anti-CD20 mAbs was similar at 100 ng/ml in
and Figure 3c shows that the cytotoxicity of NK cell-enriched CLL patients and controls (Figure 4b, lower panel). As shown in
PBMC from CLL patients was similar to that in controls. Figure 4c, in CLL patients, EC50 values were 10.5 ng/ml for
rituximab and 0.13 ng/ml for LFB-R603, which indicates that the
ADCC activity of the latter was 81 times higher than that of
ADCC potency of NK cells from CLL patients is related rituximab. In the control subjects, EC50 values were 16.2 ng/ml
to the concentration of rituximab and LFB-R603 for rituximab and 0.16 ng/ml for LFB-R603, with LFB-R603
NK-cell cytolysis can also be induced indirectly through Fc ADCC activity 101 times higher than that of rituximab. Taken
receptors, by antibodies mediating ADCC. ADCC assays with together, these results confirm the strong ADCC capacity of
purified NK cells against Raji target cells, in the presence of NK cells from CLL patients and suggest that, at least ex vivo,
serial concentrations of rituximab or LFB-R603, produced LFB-R603 is more potent than rituximab at low concentrations.
similar ADCC curves in controls and CLL patients with both The degranulation capacities of NK cells from CLL patients in
anti-CD20 mAbs, including an exponential phase followed by a the presence of their autologous B-CLL cells provide additional
plateau (Figure 4a). The effect in CLL patients was similar support for these data because ADCC is influenced not only
irrespective of their prognostic status. However, the potency by binding of NK cells to their target but also and mainly by their
was dose-dependent for both rituximab and LFB-R603, and the ability to degranulate. This ex vivo model evaluated the

Leukemia
NK cells and optimized anti-CD20 in CLL
M Le Garff-Tavernier et al
105
100 100 100

% NKG2A

% NKp30
75 75 75

% ILT-2
50 50 50
25 25 25
0 0 0
Ctl group 1 group 2 Ctl group 1 group 2 Ctl group1 group 2
100 100 100
0
75 2
% NKG2A

75 75

% NKp30

% ILT-2
4
50 50 50 6
12
25 25 25
0 0 0
#1 #2 #3 #4 #1 #2 #3 #4 #1 #2 #3 #4
CLL patients CLL patients CLL patients

* 80
80 % KIR2DL2-3/DS2 80

% KIR3DL1/DS1
% KIR2DL1/DS1

60 60 60
40 40 40
20 20 20
0 0 0
Ctl group1 group 2 Ctl group1 group 2
% CD69+/CD3-CD56bright Ctl group1 group 2

** **
% CD69+/CD3-CD56dim

* 50
75

50
25
25

0 0
Ctl group1 group 2 Ctl group1 group 2

Figure 2 Pattern of receptor expression on NK cells from CLL patients. (a) Expression of NKG2A, NKp30 and ILT-2. Upper panels show
phenotypic expression of each marker gated on CD3CD56dim NK cells. Lower panels show a longitudinal phenotypic study of four representative
CLL patients (patients 1, 2, and 4 in group 1; patient 3 in group 2), every 2 months during a 12-month period after the first phenotypic evaluation.
(b) Expression of killer cell Ig-like receptors, including KIR2DL1/DS1, KIR2DL2-3/DS2, and KIR3DL1/DS1. (c) Expression of the early
cell-activation marker CD69 on CD3CD56dim NK cells and CD3CD56bright NK cells. Cells were collected from controls (Ctl, J) and CLL
patients in group 1 (}) and group 2 (&). Horizontal bars represent the median value. P-values refer to the comparison between controls and
CLL patients, assessed with the Mann-Whitney U-test. *Po0.05, **Po0.01.

proportion of CD107a þ NK cells in whole PBMC from cells, the proportion of degranulating NK cells was considerably
CLL patients and controls. As shown in Figure 5a, analysis of higher with LFB-R603 than with rituximab.
degranulation in the presence of autologous target cells
confirmed the greater potency of LFB-R603 compared with
rituximab: although NK-cell degranulation was not observed in Discussion
samples from any of the 21 CLL patients at low concentrations
of rituximab (10 ng/ml), the median values of degranulation with This report describes the first extensive phenotypic and
the same concentration of LFB-R603 were 16.3% in group 1 and functional study of CD16 þ CD56dim NK cells in CLL patients
15.2% in group 2 (Figure 5a). Similar results were obtained and age-matched controls. CLL patients were found to have
using the Binet stages (data not shown). At a higher concentra- a significantly higher number of NK cells, as previously
tion (1000 ng/ml), NK-cell degranulation remained significantly described,22 but expression of both inhibitory and activating
higher with LFB-R603 than with rituximab (group 1, P ¼ 0.0005 NK-cell receptors was well preserved. In addition, this NK-cell
and group 2, P ¼ 0.0078) (Figure 5b). According to Binet stages, repertoire remained remarkably stable despite disease progres-
this difference remained significant in the A patient group but sion. Intriguingly, only a slight decrease in KIR2DL1/DS1
not in the B/C patient group, probably because of the low expression was noted in patients with an unfavorable prognosis,
number of patients in this latter group (data not shown). Finally, whereas the frequency of HLA-Cw*06, a KIR2DL1 ligand,
we could not exclude that CLL cells inhibited NK-cell function. was significantly higher in CLL cohorts than in controls.23,24 The
To test this hypothesis, we added Raji cells into the CD107 phenotypic features of these CD16 þ CD56dim NK cells cannot
degranulation assay performed with PBMC from CLL patients or explain the lack of antitumor activity or disease control. This
healthy controls. As shown in the Supplementary Figure 1, in the may suggest a failure to express some NK ligands on B-CLL cells,
presence of Raji cells, the CD107 level in NK cells was similar such as MICA and ULBPs, as previously reported.25 In contrast,
in CLL patients and healthy controls, suggesting an absence of the major phenotypic alterations in NK cells observed in acute
effect of CLL cells on NK degranulation. Together, these data leukemia, including the downregulation of natural cytotoxicity
demonstrate that in the presence of autologous B-CLL target receptors,26,27 suggest that chronic and acute leukemia use

Leukemia
NK cells and optimized anti-CD20 in CLL
M Le Garff-Tavernier et al
106
different strategies to escape from NK-cell immunity. Poor previously reported reduction in NK cytotoxic activity in the
expression of CD69 was also observed on CD16 þ CD56dim absence of exogenous stimulation.15,28 This relative NK-cell
NK cells from CLL patients; this deficit could explain the anergy could be due to the ability of CLL cells and their
*
microenvironment to produce cytokines, such as IL-10, which
* suppress the proliferation of antigen-specific Th1 cells and
100 100
downregulate co-stimulatory molecules on antigen-presenting
% IFN+ cells/
CD3-CD56dim

CD3-CD56bright
cells.29–31 Importantly, NK-cell function was perfectly recovered

% IFNγ+ cells/
75 75
50 50 following activation. This included the ability to produce
25 25 large amounts of IFN-g after IL-12 and IL-18 activation, and
effective cytolysis against major histocompatibility complex
0 0
Ctl group1 group 2 Ctl group1 group 2

100 10 ng/ml 1000 ng/ml


% NK lysis

75 *** ** *** **
75 75
50

+
% CD107a

% CD107a
50 50
25 42.6
35.2
0 25 25
16.3 15.2
12.4
/1

/1

/1

9.5
5/

5/
20

10

25
2.

0.0 1.4
1.

0 0
RTX LFB-R603 RTX LFB-R603 RTX LFB-R603 RTX LFB-R603
E/T ratio
group 1 group 2 group 1 group 2
Figure 3 IFN-g expression and direct cytotoxic activity of NK
cells from CLL patients. Intracellular expression of IFN-g in: Figure 5 Degranulation response determined by CD107a expression
(a) CD3CD56dim and (b) CD3CD56bright NK cells from controls on CD3CD56dim NK cells from CLL patients. PBMC from CLL
(J) and CLL patients in group 1 (}) and group 2 (&), after overnight patients in group 1 (n ¼ 12) and group 2 (n ¼ 9) were incubated
incubation with IL-12 and IL-18. Horizontal bars represent the median with: (a) 10, and (b) 1000 ng/ml of rituximab (RTX) or LFB-R603. The
values. (c) Cytotoxicity of enriched NK cells from controls (J) and median value of the % of CD107a þ NK cells without mAb was 3.5
CLL patients (~) measured by 51Cr release-assays against K562 target (range: 0.8–15.0). Horizontal black bars represent median values.
cells. Mean percentage values±s.d. (vertical bars) are shown. P-values P-values were calculated with the Wilcoxon rank-sum test and refer to
refer to the comparison between controls and CLL patients, assessed the comparison between rituximab and LFB-R603. **Po0.01,
with the Mann–Whitney U-test. *Po0.05. ***Po0.001.

1 ng/ml
* *
100 100
Ctl (n=3) n=6 n=7
% NK ADCC

% NK ADCC

75 75
62.5 65.1
50
50
25
25
0 7.3 8.5
0 0.1 1 10 100 1000 10000 0
100 ng/ml
100 n=5 n=5
100
% NK ADCC

CLL group 1 (n=4)


% NK ADCC

75 71.6
75 66.9
55.6
60.4
50
50
25 25

0 0
0 0.1 1 10 100 1000 10000 RTX LFB-R603 RTX LFB-R603
Ctl CLL
100
CLL group 2 (n=1) 100
% NK ADCC

% NK ADCC

75
75
50
50 RTX CLL
25 LFB-R603 CLL
25 RTX Ctl
0 LFB-R603 Ctl
0
0 0.1 1 10 100 1000 10000 -2 -1 0 1 2 3 4 5
[mAb] ng/mL [mAb] ng/mL (LOG)
Figure 4 NK cell ADCC in the presence of rituximab or LFB-R603. (a) A dose-response study of ADCC against Raji cells was performed with or
without anti-CD20 rituximab (n) or LFB-R603 (~), at six concentrations ranging from 0.1 to 10 000 ng/ml. Assays were performed with purified
NK cells from controls and from CLL patients in group 1 and group 2. Mean percentage values±s.d. (vertical bars) are shown. (b) Comparison of
ADCC activity against Raji cells, after treatment with 1 ng/ml or 100 ng/ml of rituximab (RTX) (n) and LFB-R603 (R603) (~). Horizontal black bars
represent median values. (c) Percentage lysis of Raji cells induced by rituximab or LFB-R603 in the presence of NK cells from control individuals
(n ¼ 3) or CLL patients (n ¼ 5). P-values were calculated with the Wilcoxon rank-sum test and refer to the comparison between rituximab and
LFB-R603, *Po0.05.

Leukemia
NK cells and optimized anti-CD20 in CLL
M Le Garff-Tavernier et al
107
class-I-defective K562 target cells after IL-2 activation. Similar the response to anti-CD20 treatment in relationship to clinical
capacities were observed in all patients, regardless of their treatment history.
prognostic factors in agreement with previous reports.17,18 In CLL, the introduction of anti-CD20 mAbs has brought
Altogether, these results contribute to our knowledge on about a remarkable change in treatment strategy, but limitations
NK-cell nature and function in CLL. in their therapeutic use have also been noted. Rituximab is not
Our results demonstrate the efficacy of NK cells from CLL widely used alone in CLL because of its low-response rate and
patients, including patients with an unfavorable prognosis, at the short-response duration. Furthermore, increased doses of
killing tumor Raji cells and degranulating against autologous rituximab, from 500 to 2250 mg/m2, are associated with
B-CLL cells. Consequently, these data strongly support the use of significant toxicity without any enhancement of clinical
anti-CD20 mAbs optimized for FcgR engagement to induce benefits.39 On the basis of these success and limitations,
efficient ADCC. Surprisingly, very few studies have evaluated different types of second-generation mAbs have been engi-
anti-CD20 mAb-mediated ADCC in CLL,9,32 with most neered. Ofatumumab (HuMax) and veltuzumab are designed to
anti-CD20 ADCC studies using non-Hodgkin’s lymphoma cell improve complement-dependent cytotoxicity, whereas GA-101
lines. Only two studies have investigated the ability of NK cells and LFB-R603, for example, are intended to increase the affinity
to mediate ADCC against primary CLL cells in the presence of of the mAb Fc region for FcgRIIIA, by amino-acid substitutions
anti-CD20 mAbs.9,32 Weitzman et al.32 showed that rituximab or modification of the carbohydrate structure of the mAb,
and veltuzumab, another anti-CD20 mAb optimized for or both. Lower doses of these antibodies must be sufficient to
enhanced complement-dependent cytotoxicity, resulted in produce ADCC levels similar to or higher than those achieved
similar levels of ADCC in the presence of primary CLL cells with rituximab, as demonstrated in this study. Clinical trials with
and NK cell lines. Recently, we have shown that EMAB-6, an these antibodies may reveal that they have enhanced activity
anti-CD20 optimized for FcgRIIIA engagement, induced greater compared with rituximab, but without its side effects. Thus, an
ADCC against CLL cells than rituximab, in the presence of NK interim analysis in an international phase I trial of ofatumumab,
cells from healthy donors.9 Of note, we focused our attention on used as single agent at a dose of 2000 mg in refractory
NK cell-mediated ADCC. However, we cannot exclude a CLL patients, reported a good safety profile and a high overall
synergistic effect of macrophages and neutrophils with NK cells response rate in heavily pretreated patients.40 A multicenter
in this mechanism.8,33 phase I study of GA-101, used as single agent at escalating doses
Other studies have focused on the cytotoxic activity of of 400–2000 mg, showed good responses with two cases of
anti-CD20 mAbs against non-Hodgkin’s lymphoma cells. With complete remission at intermediate doses (800/1200 mg), a
optimized GA-101 anti-CD20 mAb, the potency of ADCC tumor-burden decrease and good tolerance in 11/13 evaluable
against CD20-expressing non-Hodgkin’s lymphoma cells ranges CLL patients.41 Finally, an ongoing phase I study is testing
from 5 to 100 times greater than with rituximab.34 Similarly, LFB-R603 at doses escalating from 75 to 1650 mg/m2.
AME-D, another optimized anti-CD20 mAb, produces greater In summary, our findings clearly demonstrate that peripheral
ADCC at lower mAb concentrations than rituximab against an NK cells from CLL patients are functional in terms
non-Hodgkin’s lymphoma cell line.35 In the presence of NK of degranulation and ADCC, although a certain degree of
cells from CLL patients, similar dose-response relationships were variability is observed. They support the idea that a functional
found for the induction of ADCC by the optimized anti-CD20 characterization of NK cells from each CLL patient could be
LFB-R603 mAb and rituximab, except that LFB-R603 could kill useful in predicting the outcome of clinical treatment with
the same number of Raji cells at 1/100 the concentration mAbs. Our data also show that LFB-R603 activity is more
of rituximab. ADCC did not increase with concentrations higher promising than that of rituximab. The potency of LFB-R603 at
than 100 ng/ml for rituximab or 1 ng/ml for LFB-R603. Of note, low doses may have an important impact in patients with severe
LFB-R603 induces strong ADCC at a low concentration disease or in those who are resistant to chemotherapy.
(1 ng/ml), regardless of the prognostic factors.
Importantly, a robust increase in NK-cell degranulation
against autologous CLL cells was observed in the presence of Conflict of interest
LFB-R603, in contrast to the results with rituximab. Together,
these data indicate that the potency of ADCC with these two J Decocq, C de Romeuf and JF Prost are employed by LFB,
anti-CD20 mAbs differs considerably and that the anti-CD20 whose potential product was studied in the present report.
LFB-R603 mAb triggers better effector functions to clear CLL CA Dutertre was supported by a CIFRE fellowship from the
cells, which may be therapeutically beneficial. Intriguingly, ANRT and LFB (#134/2004) between 2004 and 2007. J-L
considerable interindividual variation in degranulation efficacy Teillaud was a LFB consultant until June 2007, which covers
was also observed, regardless of the patients’ prognosis. part of the study period. Three of the authors, C de Romeuf, JF
Previous studies have shown that the FcgRIIIA polymorphism Prost and JL Teillaud, are designated as inventors on patent
is associated with differential affinity and binding to human application WO2006064121 owned by LFB Biotechnologies,
IgG1 mAbs. However, in CLL, the FcgR polymorphism did not and claim specific therapeutic use of the antibody studied in this
predict responses to low-fucosylated mAbs,32,36,37 suggesting report. The other authors declare no conflict of interest.
that this polymorphism does not explain the variability in
degranulation responses. Instead, NK-cell degranulation might
be affected by the effector/target ratio, in agreement with Fischer Acknowledgements
et al.38 Finally, the individual variability might be also attributed
to intrinsic resistance of CLL cells to degranulation as observed We thank Z Azgui, K Maloum, F Nguyen-Khac, JL Binet,
in 1/21 patients. Moreover, we noted a defect in NK-cell N Marchay, H M’Kada and S Nguyen-Quoc, from the Hematology
degranulation in two patients who had received rituximab Department at Pitié-Salpêtrière Hospital (AP-HP, Paris, France) for
before sampling (data not shown). Accordingly, functional the recruitment of CLL patients. We thank F Gandjbakhch,
characterization of NK cells against autologous B-CLL targets C Poulain and the personnel from the Department of Rheuma-
from each patient might be useful for predicting the quality of tology at Pitié-Salpêtrière Hospital (AP-HP, Paris, France) and

Leukemia
NK cells and optimized anti-CD20 in CLL
M Le Garff-Tavernier et al
108
V Siguret, E Pautas and the personnel from the Gerontology 15 Ziegler HW, Kay NE, Zarling JM. Deficiency of natural killer
Department at Charles-Foix Hospital (AP-HP, Ivry, France) cell activity in patients with chronic lymphocytic leukemia.
for the recruitment of the control group. P Bonnemye, M Brissard, Int J Cancer 1981; 27: 321–327.
16 Foa R, Lauria F, Lusso P, Giubellino MC, Fierro MT, Ferrando ML
S Gueguen, M Boudjoghra and A Grelier are acknowledged for
et al. Discrepancy between phenotypic and functional features of
their technical assistance. This study was supported in part natural killer T-lymphocytes in B-cell chronic lymphocytic
by funds from the Laboratoire Français de Fractionnement et des leukaemia. Br J Haematol 1984; 58: 509–516.
Biotechnologies (LFB, Les Ulis, France), the association la Ligue 17 Kay NE, Zarling J. Restoration of impaired natural killer cell
contre le cancer (RS08/75-4) and INSERM. The anti-CD20 activity of B-chronic lymphocytic leukemia patients by recombi-
LFB-R603 antibody was purchased by LFB. nant interleukin-2. Am J Hematol 1987; 24: 161–167.
18 Guven H, Gilljam M, Chambers BJ, Ljunggren HG, Christensson B,
Kimby E et al. Expansion of natural killer (NK) and natural
Author contributions killer-like T (NKT)-cell populations derived from patients with
B-chronic lymphocytic leukemia (B-CLL): a potential source for
MLT, CAD, JLT, HMB and VV conceived and designed the study; cellular immunotherapy. Leukemia 2003; 17: 1973–1980.
MLT and JD performed the flow cytometric and functional 19 Matutes E, Owusu-Ankomah K, Morilla R, Garcia Marco J,
analyses; CP performed the flow cytometric selection; Houlihan A, Que TH et al. The immunological profile of B-cell
EC provided cytogenetic analyses; FD provided IgHV sequences; disorders and proposal of a scoring system for the diagnosis of CLL.
Leukemia 1994; 8: 1640–1645.
HMB cared for the involved patients, provided clinical and
20 Van Bockstaele F, Verhasselt B, Philippe J. Prognostic markers
diagnosis/prognosis cytometric data; MLT, JD, CdR, CAD, JLT, in chronic lymphocytic leukemia: a comprehensive review.
HMB and VV analyzed the flow cytometric and functional data; Blood Rev 2009; 23: 25–47.
MLT, JLT, HMB and VV wrote the manuscript; JD, CdR, CP, CAD, 21 Beziat V, Nguyen S, Lapusan S, Hervier B, Dhedin N, Bories D
EC, FD, PD and JFP critically read and approved the manuscript. et al. Fully functional NK cells after unrelated cord blood
transplantation. Leukemia 2009; 23: 721–728.
22 Vuillier F, Tortevoye P, Binet JL, Dighiero G. CD4, CD8 and NK
References subsets in B-CLL. Nouv Rev Fr Hematol 1988; 30: 331–334.
23 Linet MS, Bias WB, Dorgan JF, McCaffrey LD, Humphrey RL. HLA
1 Keating MJ, O’Brien S, Albitar M, Lerner S, Plunkett W, Giles F et al. antigens in chronic lymphocytic leukemia. Tissue Antigens 1988;
Early results of a chemoimmunotherapy regimen of fludarabine, 31: 71–78.
cyclophosphamide, and rituximab as initial therapy for chronic 24 Mueller LP, Machulla HK. Increased frequency of homozygosity
lymphocytic leukemia. J Clin Oncol 2005; 23: 4079–4088. for HLA class II loci in female patients with chronic lymphocytic
2 Tam CS, O’Brien S, Wierda W, Kantarjian H, Wen S, Do KA et al. leukemia. Leuk Lymphoma 2002; 43: 1013–1019.
Long-term results of the fludarabine, cyclophosphamide, and 25 Poggi A, Venturino C, Catellani S, Clavio M, Miglino M, Gobbi M
rituximab regimen as initial therapy of chronic lymphocytic et al. Vdelta1 T lymphocytes from B-CLL patients recognize ULBP3
leukemia. Blood 2008; 112: 975–980. expressed on leukemic B cells and up-regulated by trans-retinoic
3 Cartron G, Watier H, Golay J, Solal-Celigny P. From the bench to acid. Cancer Res 2004; 64: 9172–9179.
the bedside: ways to improve rituximab efficacy. Blood 2004; 104: 26 Verheyden S, Demanet C. NK cell receptors and their ligands in
2635–2642. leukemia. Leukemia 2008; 22: 249–257.
4 Lazar GA, Dang W, Karki S, Vafa O, Peng JS, Hyun L et al. 27 Costello RT, Sivori S, Marcenaro E, Lafage-Pochitaloff M,
Engineered antibody Fc variants with enhanced effector function. Mozziconacci MJ, Reviron D et al. Defective expression and
Proc Natl Acad Sci USA 2006; 103: 4005–4010. function of natural killer cell-triggering receptors in patients with
5 Delgado J, Briones J, Sierra J. Emerging therapies for patients with acute myeloid leukemia. Blood 2002; 99: 3661–3667.
advanced chronic lymphocytic leukaemia. Blood Rev 2009; 23: 28 Jewell AP, Worman CP, Giles FJ, Goldstone AH, Lydyard PM.
217–224. Resistance of chronic lymphocytic leukaemia cells to inter-
6 Pleyer L, Egle A, Hartmann TN, Greil R. Molecular and cellular feron-alpha generated lymphokine activated killer cells.
mechanisms of CLL: novel therapeutic approaches. Nat Rev Clin Leuk Lymphoma 1992; 7: 473–480.
Oncol 2009; 6: 405–418. 29 Stevenson FK, Caligaris-Cappio F. Chronic lymphocytic leukemia:
7 Cheson BD. Ofatumumab, a novel anti-CD20 monoclonal anti- revelations from the B-cell receptor. Blood 2004; 103: 4389–4395.
body for the treatment of B-cell malignancies. J Clin Oncol 2010; 30 Jak M, Mous R, Remmerswaal EB, Spijker R, Jaspers A, Yague A
28: 3525–3530. et al. Enhanced formation and survival of CD4+ CD25hi Foxp3+
8 Leidi M, Gotti E, Bologna L, Miranda E, Rimoldi M, Sica A et al. T-cells in chronic lymphocytic leukemia. Leuk Lymphoma 2009;
M2 macrophages phagocytose rituximab-opsonized leukemic 50: 788–801.
targets more efficiently than m1 cells in vitro. J Immunol 2009; 31 Pallasch CP, Ulbrich S, Brinker R, Hallek M, Uger RA,
182: 4415–4422. Wendtner CM. Disruption of T cell suppression in chronic
9 de Romeuf C, Dutertre CA, Le Garff-Tavernier M, Fournier N, lymphocytic leukemia by CD200 blockade. Leuk Res 2009; 33:
Gaucher C, Glacet A et al. Chronic lymphocytic leukaemia 460–464.
cells are efficiently killed by an anti-CD20 monoclonal antibody 32 Weitzman J, Betancur M, Boissel L, Rabinowitz AP, Klein A,
selected for improved engagement of FcgammaRIIIA/CD16. Klingemann H. Variable contribution of monoclonal antibodies
Br J Haematol 2008; 140: 635–643. to ADCC in patients with chronic lymphocytic leukemia.
10 Freud AG, Caligiuri MA. Human natural killer cell development. Leuk Lymphoma 2009; 50: 1361–1368.
Immunol Rev 2006; 214: 56–72. 33 Lefebvre ML, Krause SW, Salcedo M, Nardin A. Ex vivo-activated
11 Vivier E, Tomasello E, Baratin M, Walzer T, Ugolini S. Functions of human macrophages kill chronic lymphocytic leukemia cells in
natural killer cells. Nat Immunol 2008; 9: 503–510. the presence of rituximab: mechanism of antibody-dependent
12 Voso MT, Pantel G, Rutella S, Weis M, D’Alo F, Urbano R et al. cellular cytotoxicity and impact of human serum. J Immunother
Rituximab reduces the number of peripheral blood B-cells in vitro 2006; 29: 388–397.
mainly by effector cell-mediated mechanisms. Haematologica 34 Umana P, Moessner E, Bruenker P, Unsin G, Puentener U, Suter T
2002; 87: 918–925. et al. Novel 3rd generation humanized type II CD20 antibody with
13 Clynes RA, Towers TL, Presta LG, Ravetch JV. Inhibitory Fc glycoengineered Fc and modified elbow hinge for enhanced
receptors modulate in vivo cytoxicity against tumor targets. ADCC and superior apoptosis induction. Blood Rev 2006; 108:
Nat Med 2000; 6: 443–446. Abs 229.
14 Palmer S, Hanson CA, Zent CS, Porrata LF, Laplant B, Geyer SM 35 Bowles JA, Wang SY, Link BK, Allan B, Beuerlein G, Campbell MA
et al. Prognostic importance of T and NK-cells in a consecutive et al. Anti-CD20 monoclonal antibody with enhanced affinity for
series of newly diagnosed patients with chronic lymphocytic CD16 activates NK cells at lower concentrations and more
leukaemia. Br J Haematol 2008; 141: 607–614. effectively than rituximab. Blood 2006; 108: 2648–2654.

Leukemia
NK cells and optimized anti-CD20 in CLL
M Le Garff-Tavernier et al
109
36 Farag SS, Flinn IW, Modali R, Lehman TA, Young D, Byrd JC. blood natural killer (NK) cells. Exp Hematol 2006; 34:
Fc gamma RIIIa and Fc gamma RIIa polymorphisms do not predict 753–759.
response to rituximab in B-cell chronic lymphocytic leukemia. 39 O’Brien SM, Kantarjian H, Thomas DA, Giles FJ, Freireich EJ,
Blood 2004; 103: 1472–1474. Cortes J et al. Rituximab dose-escalation trial in chronic
37 Niwa R, Hatanaka S, Shoji-Hosaka E, Sakurada M, Kobayashi Y, lymphocytic leukemia. J Clin Oncol 2001; 19: 2165–2170.
Uehara A et al. Enhancement of the antibody-dependent 40 Wierda WG, Kipps TJ, Mayer J, Stilgenbauer S, Williams CD,
cellular cytotoxicity of low-fucose IgG1 Is independent of Hellmann A et al. Ofatumumab as single-agent CD20 immuno-
FcgammaRIIIa functional polymorphism. Clin Cancer Res 2004; therapy in fludarabine-refractory chronic lymphocytic leukemia.
10: 6248–6255. J Clin Oncol 2010; 28: 1749–1755.
38 Fischer L, Penack O, Gentilini C, Nogai A, Muessig A, Thiel E et al. 41 Morschhauser F, Cartron G, Lamy T, Milpied NJ, Thieblemont C,
The anti-lymphoma effect of antibody-mediated immuno- Tilly H et al. Phase I study of RO5072759 (GA101) in relapsed/
therapy is based on an increased degranulation of peripheral refractory chronic lymphocytic leukemia. Blood 2009: Abs 884.

Supplementary Information accompanies the paper on the Leukemia website (http://www.nature.com/leu)

Leukemia

You might also like