You are on page 1of 15

Original Article

Clinical Utility of Multiparameter Flow


Cytometry in the Diagnosis of 1013 Patients
With Suspected Myelodysplastic Syndrome
Correlation to Cytomorphology, Cytogenetics, and Clinical Data

Wolfgang Kern, MD; Claudia Haferlach, MD; Susanne Schnittger, PhD; and Torsten Haferlach, MD

BACKGROUND: The diagnosis and classification of myelodysplastic syndromes (MDS) is based on cytomorphology
(CM) and cytogenetics (CG). Multiparameter flow cytometry (MFC) may add important diagnostic information.
METHODS: To evaluate the potential role of MFC in the diagnostic setting of MDS, the authors analyzed the results
from 1013 patients with suspected MDS by using CM, CG, and MFC in parallel. RESULTS: Concordance between CM
and MFC was 82% for diagnostic results in 788 patients who had unequivocal CM results. An additional 225 patients
had only minor dysplastic features identified by CM, including 51 patients (22.7%) who had clear evidence of MDS by
MFC. Twelve patients who had no indication of MDS identified by CM had MDS-typical CG aberrations; in 6 of those
patients (50%), MFC revealed MDS characteristics. In another 11 of 23 patients (47.8%) who had minor dysplastic fea-
tures identified by CM and MDS-typical CG aberrations, MFC revealed MDS characteristics. The percentages of blasts
determined by CM and by MFC were strongly correlated (P < .001). The frequency of aberrantly expressed antigens
differed significantly between patients rated by CM as MDS (highest frequencies), suspected MDS, and no MDS (low-
est frequencies). In various patients, MFC identified MDS-typical aberrant antigen expression in cell compartments
that were not rated dysplastic by CM. The numbers of aberrantly expressed antigens were correlated with Interna-
tional Prognostic Scoring System scores and overall survival. CONCLUSIONS: The current analysis clearly demon-
strated an increased diagnostic yield with MFC when added to CM and CG in patients with suspected MDS. Cancer
2010;116:4549–63. V C 2010 American Cancer Society.

KEYWORDS: myelodysplasia, flow cytometry, cytomorphology, cytogenetics.

Myelodysplastic syndromes (MDS) comprise a heterogeneous group of clonal diseases that originate in malig-
nant hematopoietic stem cells.1 Clinical manifestations of MDS result from peripheral blood cytopenias and include fa-
tigue, infections, and bleeding.1 The diagnosis of MDS relies on the evaluation of dysplastic findings in bone marrow
(BM) cells. Acquired chromosomal aberrations are detected in 50% of patients.2-4
Diagnostic criteria and classification of MDS have been based on the French-American-British (FAB) classification
for 2 decades5 with standardized criteria for dysplasia.6 Cytogenetic (CG) analysis has been included in the standard diag-
nostic workup and classification.7-9 In addition to cytomorphology (CM) and CG, the number of cytopenias and transfu-
sion requirements have been included in the powerful International Prognostic Scoring System (IPSS)10 and World
Health Organization (WHO) classification-based Prognostic Scoring System,11 respectively, for prognosis.
It has been demonstrated that both FAB and the WHO classification identify clinically relevant subgroups.12 In
patients with chromosomal aberrations, CG may contribute substantially to establish a diagnosis of MDS if CM results
are equivocal. However, in a large number of patients with cytopenias, the diagnosis may not be made, and repeat evalua-
tions are recommended.
Because of these diagnostic difficulties, it is worthwhile to test the power of multiparameter flow cytometry (MFC).
MFC repeatedly has demonstrated the ability to identify the aberrant expression of antigens that are considered dysplastic

Corresponding author: Wolfgang Kern, MD, MLL Munich Leukemia Laboratory, Max-Lebsche-Platz 31, 81377 Munich, Germany; Fax: (011) 49-89-990-17-209;
wolfgang.kern@mll-online.com
MLL Munich Leukemia Laboratory, Munich, Germany
DOI: 10.1002/cncr.25353, Received: October 5, 2009; Revised: February 21, 2010; Accepted: February 25, 2010, Published online June 22, 2010 in Wiley Online
Library (wileyonlinelibrary.com)

Cancer October 1, 2010 4549


Original Article

Table 1. Panel of Monoclonal Antibodies Used for Immunophenotyping

Combination FITC PE ECD PC5 PC7


1 CD11b CD13 HLA-DR CD16 CD45
MoAb Bear1 Immu103.44 Immu357 3G8 J33

2 CD71 CD235a CD19 CD2 CD45


MoAb YDJ1.2.2 11E4B-7-6 J3-119 39C1.5 J33

3 CD61 CD5 CD14 CD4 CD45


MoAb SZ21 BL1A RMO52 13B8.2 J33

4 CD15 CD66c CD3 CD64 CD45


MoAb 80H5 B6.2 UCHT1 22 J33

5 CD65 CD56 CD10 CD33 CD45


MoAb 88H7 N901 ALB1 D3HL60.251 J33

6 CD36 CD38 CD34 CD7 CD45


MoAb FA6-152 T16 581 8H8.1 J33

FITC indicates fluorescein isothiocyanate; PE, phycoerythrin; ECD, phycoerythrin-Texas Red; PC5, phycoerythrin-cyanine 5; PC7, phycoerythrin-cyanine 7;
MoAb, monoclonal antibody; CD, cluster of differentiation; CD11b, integrin a M; CD13, alanine aminopeptidase; HLA-DR, human leukocyte antigen-D related;
CD16, human neutrophil antigen 1; CD45, protein tyrosine phosphatase, receptor type, C; CD71, transferrin receptor 1; CD235a, glycophorin A; CD19, protein
encoded by the CD19 B-lymphocyte antigen gene; CD2, lymphocyte function-associated antigen 2; CD61, protein encoded by the integrin beta 3 gene; CD5,
type I transmembrane protein; CD14, monocyte differentiation antigen CD14; CD4, protein encoded by the T-cell surface glycoprotein CD4 gene; CD15, 3-
fucosyl-N-acetyl-lactosamine; CD66c, carcinoembryonic antigen-related cell adhesion molecule 6 (nonspecific cross-reacting antigen); CD3, T-cell coreceptor;
CD64, integral membrane glycoprotein; CD65, fucosylated carbohydrate cell-surface antigen; CD56, neural cell adhesion molecule; CD10, common acute lym-
phocytic leukemia antigen; CD33, 67-kDa membrane glycoprotein; CD36, collagen type 1 receptor; CD38, cyclic adenine diphosphate hydrolase; CD34, cell
surface antigen, glycoprotein 105-120; CD7, glycoprotein 40.

features.13-16 These findings have been described in MDS 4.0-20.2 g/dL); and median thrombocyte count, 157,500/
and do not occur or occur only infrequently in patients lL, (range, 2000-1195,000/lL).
who have cytopenias that are not considered MDS. The
objective of the current study was to estimate the potential Cytomorphology and Cytogenetics
diagnostic value of MFC. CM assessment was based on May-Grunwald-Giemsa
stains, myeloperoxidase reactions, and nonspecific esterase
using alpha-naphtyl-acetate17-19 and was used for diagno-
MATERIALS AND METHODS sis according to FAB20-22 and WHO7,8 criteria. CG anal-
Patients yses were performed according to standard protocols.
In total, 1013 BM samples from patients with suspected Classification was according to the International System
MDS were sent to MLL Munich Leukemia Laboratory for Human Cytogenetic Nomenclature.23-25 Complex
between August 2005 and January 2009 and were ana- aberrant karyotypes were defined by 3 clonal chromo-
lyzed in parallel by using CM, CG, and MFC. The suspi- some aberrations.8,26 Fluorescence in situ hybridization
cion of MDS was raised when the physician sent the was used according to standard procedures27 to clarify all
sample and was based on peripheral blood count and dif- difficult cases.
ferential as well as clinical criteria. Patients who had
hematologic malignancies other than MDS were excluded Multiparameter Flow Cytometry
after the respective diagnosis was confirmed. Thus, in the Samples were processed by Ficoll gradient centrifugation
current cohort, all patients had in common an initial suspi- followed by ammonium chloride-based erythrocyte lysis to
cion of MDS. All 3 diagnostic methods were performed in- isolate mononuclear cells. MFC was performed by applying
dependently from each other. CM was performed by T.H., 5-fold stainings and using the antibodies outlined in Table
CG was performed by C.H., and MFC was performed by 1 (selected based on previous studies).13-15,28,29 Antibodies
W.K. The median patient age was 69.8 years (range, 1.8- were purchased from Immunotech (Marseilles, France)
88.7 years), and the ratio of men to women was 539:474. except for carcinoembryonic antigen-related cell adhesion
Median values and ranges for peripheral blood counts were molecule 6/nonspecific cross-reacting antigen (cluster of
as follows: median white blood cell count, 4400/lL (range, differentiation [CD] 66c [CD66c]) (Becton Dickinson,
600-117,600/lL); median hemoglobin, 10.7 g/dL (range, Heidelberg, Germany). Antibody combinations were

4550 Cancer October 1, 2010


MFC in MDS/Kern et al

Figure 1. Cluster of differentiation 13 (alanine aminopeptidase) (CD13)/human neutrophil antigen (CD16) expression patterns are
shown (Left) in granulocytes from normal bone marrow (BM) and (Right) in myelodysplastic syndrome (MDS) PC5 indicates phy-
coerythrin-cyanine 5; PE, phycoerythrin.

added to 106 mononuclear cells (volume, 100 lL) and gene (CD19)-positive B-lymphoid progenitors, and in
incubated for 10 minutes. After the addition of 2 mL lys- addition, were demonstrated to express cell surface anti-
ing solution, samples were incubated for additional 10 gen-glycoprotein 105-120 (CD34), or human leukocyte
minutes, then washed twice in phosphate-buffered saline antigen-D related (HLA-DR), or both. The percentage of
(PBS), and resuspended in 0.5 mL PBS. FC500 flow blasts was calculated by using the total amount of mono-
cytometers were used (Beckman Coulter, Miami, Fla), nuclear cells after sample preparation as the denominator.
and 20,000 events were acquired. Cytomics CXP Soft- According to previous reports,13-15,28,29 the following fea-
ware (Beckman Coulter) was used for data analysis. tures were evaluated.
Granulocytes were evaluated for SSC signal, abnormal
Gating Strategy alanine aminopeptidase (CD13)/human neutrophil antigen
Protein tyrosine phosphatase, receptor type, C (CD45)- 1 (CD16) and integrin a M (CD11b)/CD16 expression
side scatter (SSC) gating was performed to identify cellu- patterns, neural cell adhesion molecule (CD56) coexpres-
lar compartments to allow the separate evaluation of anti- sion, 67-kDa membrane glycoprotein (CD33) negativity,
gen expression for each compartment.30 The lack of and integral membrane glycoprotein (CD64) negativity.
expression of an antigen is indicated by ‘‘,’’ dim expres- The CD13/CD16 expression pattern was considered
sion is indicated by ‘‘(þ),’’ any expression is indicated by abnormal if a deviation of normal granulocytic matura-
‘‘þ,’’ and strong expression is indicated by ‘‘þþ.’’ A tion (CD13þ/CD16!CD13[þ]/CD16!CD13/
broader range of expression is indicated by 2 respective CD16[þ]!CD13/CD16þ!CD13þ/CD16þþ) was
values separated by a virgule, ie, expression ranging from observed (Fig. 1). Also, the CD11b-/CD16 expression
‘‘(þ)’’ to ‘‘þ’’ is indicated by ‘‘(þ)/þ.’’ Granulocytes were pattern was considered abnormal if a deviation of normal
identified as SSCþ/(þ)CD45(þ), monocytes were iden- granulocytic maturation (CD11b/CD16!CD11bþ/
tified as SSC(þ)CD45þ, myeloid blasts were identified CD16[þ]!CD11bþ/CD16þ!CD11bþþ/CD16þþ)
as SSC([þ])CD45(þ), and erythroid cells were identified was observed. Deviation from normal was defined as a dif-
as CD45 glycophorin A (CD235a)þþ. It should be ference that amounted to at least a half-log signal intensity
recognized that, because a DNA-binding dye was not in 1 parameter, although it must be recognized that the
used, there is no guarantee that the erythroid cells identi- assessment is always at least partially subjective.
fied are truly nucleated. By applying back-gating, mono- Monocytes were evaluated for CD11b negativity,
cytes also were identified as monocyte differentiation HLA-DR negativity, CD13 negativity, CD16 coexpres-
antigen CD14 (CD14)þ, and blasts were separated from sion, CD56 coexpression, aberrant lymphocyte function-
the protein encoded by the CD19 B-lymphocyte antigen associated antigen 2 (CD2) coexpression.

Cancer October 1, 2010 4551


Original Article

Figure 2. (Left) Monocytes in normal bone marrow (BM) revealed no expression of cluster of differentiation 56 (CD56) (neural
cell adhesion molecule). (Right) In myelodysplastic syndrome (MDS), strong CD56 expression was observed. CD33 indicates 67-
kDa membrane glycoprotein; PC5, phycoerythrin-cyanine 5; PE, phycoerythrin.

Figure 3. (Left) In normal bone marrow (BM), the majority of erythrocytes had strong expression of cluster of differentiation 71
(CD71) (transferrin receptor 1) with some degree of heterogeneity for dim expression. (Right) In myelodysplastic syndrome
(MDS), negative CD71 expression may be present in the majority of erythrocytes. CD235a indicates glycophorin A; PE, phycoery-
thrin; FITC, fluorescein isothiocyanate.

Myeloid blasts spective isotype controls using 20% as the cutoff. The
Myeloid blasts were evaluated for the percentage of SSC signal in granulocytes was assessed qualitatively as ei-
BM myeloid blasts, coexpression of CD11b, type I trans- ther reduced or not reduced based on the CD45/SSC dot
membrane protein (CD5), CD56, transferrin receptor plot. For a marker with which to describe objectively the
1(CD7), CD15, and CD64 and HLA-DR negativity. reduced SSC signal in granulocytes, the granulocyte-to-
Erythroid cells
lymphocyte ratio of the mean SSC signal as acquired on a
Erythoroid cells were evaluated for homogeneously linear scale was calculated for each case (the SSC granulo-
strong transferrin receptor 1 (CD71) expression and cyte:lymphocyte ratio).
CD71 negativity. Examples of aberrant antigen expres-
sion are shown in Figures 1, 2, and 3. Statistical Analysis
Positivity and negativity, respectively, of the respec- Dichotomous variables were compared using chi-square
tive antigens were determined by comparison with the re- tests, and continuous variables were analyzed with the

4552 Cancer October 1, 2010


MFC in MDS/Kern et al

Table 2. Diagnostic Results of Multiparameter Flow Table 3. Diagnostic Results of Multiparameter Flow
Cytometry in Cytomorphologically Defined Myelodysplastic Cytometry in Cytogenetically Defined Myelodysplastic
Syndrome Subgroups Syndrome Subgroups

CM Total No. of Cytogenetic Result Total No. of


Result No. of Patients With No. of Patients With
Patients Results in Patients Results in
Agreement With Agreement With
MDS by MFC MDS by MFC
Independent of Independent of
CM Results (%) CM Result (%)
RA 31 22 (71) Normal karyotype 768 257 (33.5)
RARS 27 16 (59.3) Del(5q) 43 33 (76.7)
RCMD 64 23 (35.9) Aberrations of chromosome 7 14 14 (100)
RCMD-RS 49 32 (65.3) Trisomy 8 30 25 (83.3)
RAEB-1 133 104 (78.2) Del(20q) 21 18 (85.7)
RAEB-2 81 78 (96.3) Complex karyotype 23 19 (82.6)
5q– Syndrome 24 18 (75) Loss of Y-chromosome 43 22 (51.2)
CMML 65 62 (95.4) Other aberrations 71 58 (81.7)
MDS-u 15 12 (80)
MDS/AML 6 6 (100) MFC indicates multiparameter flow cytometry; MDS, myelodysplastic
MDS/MPS 16 9 (56.3) syndrome; CM, cytomorphology; Del, deletion.
Suspected MDS 225 51 (22.7)
Reactive condition 266 13 (4.9)
Normal findings 11 0 (0) lowest percentages of results that were in agreement
CM indicates cytomorphology; MDS, myelodysplastic syndrome; MFC, multi- with MDS by MFC were observed in the MDS sub-
parameter flow cytometry; RA, refractory anemia; RARS, refractory anemia group of patients who had refractory cytopenia with
with ring sideroblasts; RCMD, refractory cytopenia with multilineage dysplasia;
RCMD-RS, refractory cytopenia with multilineage dysplasia and ring sidero-
multilineage dysplasia (RCMD); however, the percen-
blasts; RAEB-1, refractory anemia with 5% to 9% excess blasts; RAEB-2, re- tages still clearly were higher than the respective per-
fractory anemia with 10% to 19% excess blasts; , negative; CMML, chronic
myelomonocytic leukemia; MDS-u; unclassifiable myelodysplastic syndrome;
centages observed in patients with suspected MDS
AML, acute myeloid leukemia; MPS, myeloproliferative syndrome. according to CM results and in patients with no
MDS. CG results are detailed in Table 3. Fluores-
Student t test. Spearman rank correlation was used to ana-
cence in situ hybridization was performed in 443
lyze correlations between continuous parameters. Survival
cases.27
was analyzed using the Kaplan-Meier method,31 and differ-
ences were analyzed using the log-rank test. All calculations
were performed using SPSS software (version 14.0.1; SPSS
Comparison of Diagnostic Results Obtained
Inc., Chicago, Ill). All reported P values are 2-sided. by Cytomorphology and Multiparameter
Flow Cytometry
Study Conduct There was 82% percent concordance between CM and
Patients provided informed consent to participate in the MFC for diagnostic results (646 of 788) in patients with
current diagnostic procedures and the evaluation at MLL definite CM results. In detail, 511 patients were classified
Munich Leukemia Laboratory after they were advised with MDS according to CM, including 382 patients
about the purpose and investigational nature of the study (74.8%) who had antigen expression features according to
and of the potential risks. The study design adhered to the MFC in agreement with MDS. Of 277 patients who had
Declaration of Helsinki. CM results indicating no MDS, only 13 patients (4.7%)
had MDS-typical features according to MFC. The pattern
of these MDS-typical features did not differ from the
RESULTS overall pattern observed in the total cohort. Two hundred
Cytomorphologic and Cytogenetic twenty-five patients had some dysplastic features on CM
Characterization but did not fulfill the diagnostic criteria for MDS; how-
Classification by CM is detailed in Table 2. The ever, 51 of those patients (22.7%) had MDS-typical find-
highest percentages of results that were in agreement ings on MFC. Clinical follow-up of these patients is not
with MDS by MFC were observed in the MDS sub- available to an extent that would allow reasonable clinical
groups with blast counts >10%, as anticipated. The validation.

Cancer October 1, 2010 4553


Original Article

Comparison of Cytogenetics and Diagnostic evidence for MDS on CM, and 50% of those patients had
Results Obtained by Multiparameter Flow an aberrant karyotype. In addition, 5.1% of all patients had
Cytometry MFC results that were in agreement with MDS, whereas
Two hundred forty-five patients had an aberrant karyo- the CM results indicated suspected MDS, and slightly less
type, and 189 (77.1%) of those patients had MDS accord- than 33% of those patients had an aberrant karyotype.
ing to the MFC results (Table 3). Thus, 6.4% of all patients had MFC results that were in
agreement with MDS without a clear diagnosis of MDS by
Comparison of Diagnostic Results Obtained CM, and 33% of those patients had an aberrant karyotype.
by Multiparameter Flow Cytometry,
Cytomorphology, and Cytogenetics
Multiparameter Flow Cytometry in
The numbers of patients who had results that were rated in
Cytogenetically Aberrant Cases Not
agreement and not in agreement with MDS on MFC are Classified as Myelodysplastic Syndrome by
listed separately according to the CM results (no MDS, Cytomorphology
MDS, or suspected MDS), and each category is broken Loss of the Y chromosome was not considered MDS-related,
down according to aberrant CG results and normal CG because it occurs in healthy, older individuals. Thus,
results in Table 4. Overall, 1.3% of patients had MFC although these patients are listed in Table 5, they are not
results that were in agreement with MDS in the absence of included in the totals. In 12 patients, CM gave no indication
Table 4. Diagnostic Results of Multiparameter Flow of MDS, but MDS-typical CG aberrations were present (Ta-
Cytometry in Cytomorphologically Defined Myelodysplastic
Syndrome Subgroups Separated According to the Presence
ble 5). The MFC results from 6 of 12 patients (50%) were in
of Cytogenetic Abnormalities agreement with MDS. In another 23 patients, CM identified
dysplastic features that were not sufficient to diagnose MDS,
MFC Result in
Agreement With MDS but CG revealed aberrations. MFC results from 11 of 23
Independent of patients (47.8%) were in agreement with MDS. It is note-
CM Result: No. of worthy that patients who had loss of the Y chromosome had
Patients (%)
MFC results that were in agreement with MDS only if CM
CM Result No Yes
identified dysplastic features that were not sufficient to diag-
Aberrant cytogenetics nose MDS, whereas all patients who had loss of the Y chro-
No MDS 17 (1.7) 6 (0.6)
mosome and no indication of MDS on CM had MFC
MDS 22 (2.2) 168 (16.6)
Suspected MDS 17 (1.7) 15 (1.5) results that were not in agreement with MDS (Table 5).
Normal cytogenetics
No MDS 247 (24.4) 7 (0.7) Comparison of Myeloid Blasts Counts by
MDS 107 (10.6) 214 (21.1) Cytomorphology and Multiparameter Flow
Suspected MDS 157 (15.5) 36 (3.6) Cytometry
MFC indicates multiparameter flow cytometry; MDS, myelodysplastic syn- The mean  standard deviation percentages of myeloid
drome; CM, cytomorphology. blasts determined by CM versus MFC were 4.67% 
Table 5. Diagnostic Results of Multiparameter Flow Cytometry in Patients With Cytogenetically
Aberrant Results Not Classified as Myelodysplastic Syndrome by Cytomorphology

Suspected MDS by CM No MDS by CM


Cytogenetic Result Total No. No. of Patients Total No. No. of Patients
of Patients With Results in of Patients With Results in
Agreement With Agreement With
MDS by MFC MDS by MFC
Del(5q) 5 3 1 0
Aberrations of chromosome 7 1 1 3 3
Trisomy 8 3 1 3 1
Del(20q) 4 3 1 0
Complex karyotype 1 0 1 1
Loss of Y chromosome 5 4 10 0
Other aberrations 9 3 3 1

MDS indicates myelodysplastic syndrome; CM, cytomorphology; MFC, multiparameter flow cytometry; Del, deletion.

4554 Cancer October 1, 2010


MFC in MDS/Kern et al

Figure 4. The correlation of blast counts obtained by cyto-


morphology (CM) and by multiparameter flow cytometry
(MFC) is depicted. The height of each column indicates the
numbers of patients. The mean  standard deviation percen-
tages of myeloid blasts determined by CM versus MFC were
4.67%  4.18% versus 3.78%  2.97%, respectively (Spearman
rank correlation, 0.362; P < .001).

4.18% versus 3.78%  2.97% (Spearman rho [r], 0.362;


P < .001) (Fig. 4). A higher blast count on CM in some
patients resulted from the presence of monocytoid cells in
which monocytic characteristics were featured that had to
be included in the blast counts according to WHO criteria
but that did not express progenitor markers according to
the MFC results. However, these cells clearly were mye-
loid/monoblastic or promonocytic blasts that were
detected by CM and cytochemistry (for an example, see
Fig. 5). Accordingly, in patients who had aberrant CD56
expression on monocytes, a higher blast count (as deter-
mined by CM) was observed compared with patients who
did not have aberrant CD56 expression (6.1%  4.6% vs
4.1%  3.9%; P < .001), whereas the respective differen-
ces in blast counts determined by MFC clearly were
smaller (4.1%  3.2% vs 3.6%  2.9%; P ¼ .014).

Characteristics in Patients With Figure 5. A bone marrow sample from a patient with myelo-
Myelodysplastic Syndromes Defined by dysplastic syndrome is shown in which multiparameter flow
Multiparameter Flow Cytometry cytometry yielded (A) a myeloid blast count of 6% (red) and
(A,B) 14% monocytic cells (purple), whereas (C) cytomor-
Aberrant antigen expression in myeloid blasts phology yielded a myeloid blast count of 18%. SSC indicates
Aberrant antigen expression in myeloid blasts was side scatter; CD, cluster of differentiation; CD45, protein tyro-
sine phosphatase, receptor type, C; PC7, proprotein conver-
most frequent in patients who had MDS compared with tase 7; CD33, 67-kDa membrane glycoprotein; PC5,
patients who did not have MDS or who had suspected phycoerythrin-cyanine 5; CD56, neural cell adhesion mole-
MDS according to the CM results (Table 6). In patients cule; PE, phycoerythrin.

who had CG aberrations, an aberrant expression of the

Cancer October 1, 2010 4555


Original Article

Table 6. Correlation of Aberrant Antigen Expression in Myeloid Blasts With Blast Count and
Dysplastic Features in Cytomorphologya

Cytomorphologic Findings: No. of


Patients (%)
MFC Findings No MDS, MDS, Suspected P
n5277 n5511 MDS, n5225
CD11bþ 3 (1.1) 27 (5.3) 13 (5.8) .009
HLA-DR 0 (0) 4 (0.8) 0 (0) NS
CD5þ 0 (0) 9 (1.8) 0 (0) .012
CD56þ 0 (0) 17 (3.3) 4 (1.8) .007
CD7þ 1 (0.4) 18 (3.5) 1 (0.4) .002
CD15þ 1 (0.4) 10 (2) 6 (2.7) NS
CD64þ 1 (0.4) 12 (2.3) 6 (2.7) NS
No. of aberrant antigens 0.00.2b,c 0.20.6b 0.10.6c .001b
per patient, meanSD .003c
% Blasts, meanSD 2.81.6b,c 4.63.7b 3.11.7c .001b
.041c

MFC indicates multiparameter flow cytometry; MDS, myelodysplastic syndrome; CD, cluster of differentiation;
CD11b, integrin a M; þ, positive; HLA-DR, human leukocyte antigen-D related; , negative; NS, nonsignificant; CD5,
type I transmembrane protein; CD56, neural cell adhesion molecule; CD7, glycoprotein 40; CD15, 3-fucosyl-N-acetyl-
lactosamine; CD64, integral membrane glycoprotein; SD, standard deviation.
a
Numbers of patients were compared using chi-square tests, and mean values were determined using Student t tests.
b
Comparison of columns 1 and 2.
c
Comparison of columns 1 and 3.

following antigens was observed more frequently com- (Table 7). Aberrant antigen expression was not correlated
pared with their expression in patients who had normal strongly with dysgranulopoiesis by CM. Thus, in 406
CG results: CD11b (7.3% vs 3.3%; P ¼ .010), CD56 patients without dysgranulopoiesis according to CM
(4.1% vs 1.4%; P ¼ .018), and CD7 (3.7% vs 1.4%; results, dysplastic features were observed with the follow-
P ¼ .036). ing frequencies: aberrant CD13/CD16, 104 patients
Considering CG subgroups, a blast count >5% was (25.6%); aberrant CD11b/CD16, 62 patients (15.3%);
observed more often (compared with the remaining CD56 expression, 38 patients (9.4%); CD33 negativity,
patients) in those who had aberrations of chromosome 7 44 patients (10.8%); and CD64 negativity, 2 patients
and in those who had a complex aberrant karyotype, (0.5%). Aberrant expression of 2 antigens in granulo-
respectively (7 of 14 patients [50%] vs 167 of 999 patients cytes was observed in 16 of 31 patients (51.6%) and in 15
[16.7%]; P ¼ .005 and 14 of 23 patients [60.9%] vs 160 of 27 patients (55.6%) with refractory anemia (RA) and
of 990 patients [16.1%]; P < .001). Accordingly, the RA with ring sideroblasts (RARS), respectively.
mean (standard deviation) blast counts were higher in The SSC granulocyte:lymphocyte ratio was lower in
the former patients (5.8  3.3 vs 3.8  3.0; P ¼ .039 and patients with MDS than in patients with no MDS
8.3  5.5 vs 3.7  2.8; P < .001). Patients with 20q dele- (6.55  2.32 vs 7.47  1.09, respectively; P < .001). Fur-
tion (del[20q]) or del(5q), respectively, as the sole aberra- thermore, no respective significant differences were
tion had CD11b expression more often than others (4 of observed between patients with or without cytomorpho-
21 patients [19%] vs 39 of 992 patients [3.9%], respec- logically identified dysgranulopoiesis (6.99  2.20 vs
tively; P ¼ .010; and 5 of 43 patients [11.6%] vs 38 of 6.98  1.27, respectively; P value not significant.).
970 patients [3.9%], respectively; P ¼ .010). In patients who had CG aberrations, aberrant
expression of the following antigens was observed more
Aberrant antigen expression in granulocytes frequently compared with patients who had a normal
In granulocytes, aberrant antigen expression was karyotype: CD13/CD16 (42.9% vs 25.1%, respectively;
observed most frequently in patients who had MDS con- P < .001), CD11b/CD16 (24.1% vs 15.4%, respectively;
firmed by CM compared with patients who had no MDS P ¼ .003), CD56 (20.8% vs 9.4%, respectively; P <
or who had suspected MDS according to the CM results .001), CD33 negativity (12.7% vs 7.7%, respectively;

4556 Cancer October 1, 2010


MFC in MDS/Kern et al

Table 7. Correlation of Aberrant Antigen Expression in Granulocytes With Cytomorphologya

Cytomorphologic Findings: No. of


Patients (%)
MFC Findings No MDS, MDS, Suspected P
n5277 n5511 MDS, n5225
Abnormal CD13/CD16 25 (9) 219 (42.9) 54 (24) <.001
Abnormal CD11b/CD16 9 (3.2) 143 (28) 25 (11.1) <.001
CD56þ 10 (3.6) 90 (17.6) 23 (10.2) <.001
CD33 18 (6.5) 53 (10.4) 19 (8.4) NS
CD64 0 (0) 14 (2.7) 8 (3.6) .011

No. of aberrant antigens 0.00.2


per patient, meanSD
1 0.20.6 <.001
2 0.10.6 .003
Reduced SSC signal 14 (5.1) 286 (56) 42 (18.7) <.001

SSC ratio of granulocytes 7.471.09


to lymphocytes, meanSD
1 6.552.32 <.001
2 7.381.17 NS

MFC indicates multiparameter flow cytometry; MDS, myelodysplastic syndrome; CD, cluster of differentiation; CD13,
alanine aminopeptidase; CD16, human neutrophil antigen 1; CD11b, integrin a M; CD56, neural cell adhesion mole-
cule; þ, positive; CD33, transmembrane receptor; , negative; CD64, integral membrane glycoprotein; NS, nonsignifi-
cant; SD, standard deviation; SCC, side scatter.
a
Numbers of patients were compared using chi-square tests, and mean values were determined using Student t tests.

P ¼ .020), and CD64 negativity (4.9% vs 1.3%, respec- 7.02  1.89 without trisomy 8 [P < .001]; 20q alone,
tively; P ¼ .002). Patients who had aberrant CG results 6.25  1.13 vs 7.00  1.89 without 20q alone
had a lower SSC granulocyte:lymphocyte ratio [P ¼ .007]; 5q alone, 6.03  1.20 vs 7.03  1.90 with-
(6.33  1.26 vs 7.20  2.00, respectively; P < .001). out 5q alone [P < .001]; and a complex aberrant karyo-
In patients who had aberrations of chromosome 7 type, 5.83  1.28 vs 7.01  1.89 without a complex
only and in patients who had complex aberrant karyo- aberrant karyotype [P < .001]).
types, distinct aberrant antigen expression was observed
more frequently (for this comparison and for the compari- Aberrant antigen expression in monocytes
sons described below, the remaining patients were used as In patients who had both MDS and suspected MDS
comparison groups): CD13/CD16, 9 of 14 patients according to CM results, an aberrant antigen expression in
(64.3%) versus 289 of 999 patients (28.9%), respectively monocytes was observed more frequently compared with
(P ¼ .007) and 12 of 23 patients (52.2%) versus 286 of patients who had no MDS (Table 8). CD56 coexpression
990 patients (28.9%), respectively, (P ¼ .020). was observed most often in patients with MDS (43.1%);
Patients who had 5q alone also had other specific however, it also was encountered in patients without CM
markers: CD56 was present in 10 of 43 patients (23.3%) evidence of MDS (9.4%). Similar percentages were
versus 113 of 970 patients without 5q (11.6%; observed for CD16 expression (9% vs 3.6%, respectively),
P ¼ .031). Among those with trisomy 8, aberrant CD13/ whereas both CD2 coexpression (8.8% vs 0.4%, respec-
CD16 expression was observed in 16 of 30 patients tively) and CD13 negativity (8.6% vs 2.2%, respectively)
(53.3%) versus 282 of 983 patients without trisomy 8 clearly occurred more often in patients with MDS.
(28.7%; P ¼ .007); aberrant CD11b/CD16 expression The following analyses were made using the respec-
was observed in 13 of 30 patients (43.3%) versus 164 of tive remaining patients in each subgroup for comparisons.
983 patients (16.7%), respectively (P ¼ .001); and CD33 Aberrantly expressed antigens in monocytes were
negativity was observed in 9 of 30 patients (30%) versus observed in patients with chronic myelomonocytic leuke-
81 of 983 patients (8.2%), respectively (P ¼ .001). Fur- mia (CMML) more frequently (vs patients without
thermore, the following CG subgroups had a lower SSC CMML), including CD13 negativity in 9 of 65 patients
granulocyte:lymphocyte ratios (trisomy 8, 6.00  1.08 vs (13.8%) versus 54 of 948 patients (5.7%; P ¼ .015),

Cancer October 1, 2010 4557


Original Article

Table 8. Correlation of Aberrant Antigen Expression in Monocytes With Cytomorphologya

Cytomorphologic Findings: No. of


Patients (%)
MFC Findings No MDS, MDS, Suspected P
n5277 n5511 MDS, n5225
CD11b 1 (0.4) 7 (1.4) 3 (1.3) NS
HLA-DR 3 (1.1) 44 (8.6) 6 (2.7) <.001
CD13 6 (2.2) 44 (8.6) 13 (5.8) .002
CD16þ 10 (3.6) 46 (9) 15 (6.7) .018
CD56þ 26 (9.4) 220 (43.1) 47 (20.9) <.001
CD2þ 1 (0.4) 45 (8.8) 6 (2.7) <.001

No. of aberrant antigens 0.20.5 <.001


per patient, meanSD
1 0.80.9
2 0.40.7

MFC indicates multiparameter flow cytometry; MDS, myelodysplastic syndrome; CD, cluster of differentiation;
CD11b, integrin a M; , negative; NS, nonsignificant; HLA-DR, human leukocyte antigen-D related; CD13, alanine
aminopeptidase; CD16, human neutrophil antigen 1; þ, positive; CD56, neural cell adhesion molecule; CD2, lympho-
cyte function-associated antigen 2; SD, standard deviation.
a
Numbers of patients were compared with chi-square tests, and mean values were determined with Student t tests.

CD56 expression in 53 of 65 patients (81.5%) versus 240 In patients with CG aberrations (vs patients without
of 948 patients (25.3%; P < .001), and CD2 expression CG aberrations), CD56 expression was observed more fre-
in 14 of 65 patients (21.5%) versus 38 of 948 patients quently (95 of 245 patients [38.8%] vs 198 of 768 patients
(4%; P < .001). [25.8%]; P < .001). In patients who had MDS with a com-
Fifteen of 133 patients (11.3%) who had RA with plex aberrant karyotype (vs patients who had MDS with nor-
excess blasts (RAEB) between 5% and 9% (RAEB-1) were mal CG results), HLA-DR negativity was observed in 5 of
negative for HLA-DR compared with 48 of 880 patients 23 patients (21.7%) versus 48 of 909 patients (4.8%), respec-
without RAEB-1 (5.5%; P ¼ .019); CD13 negativity was tively (P ¼ .005). In patients who had MDS with trisomy 8
observed in 15 of 133 patients (11.3%) versus 38 of 880 (vs patients who had MDS without trisomy 8), CD13 nega-
patients (4.3%), respectively (P ¼ .020); CD16 expres- tivity was observed more frequently (7 of 30 patients
sion was observed in 16 of 133 patients (12%) versus 55 [23.3%] vs 56 of 983 patients [5.7%]; P ¼ .002), and CD56
of 880 patients (6.3%), respectively (P ¼ .027); and expression was observed more frequently (19 of 30 patients
CD56 expression was observed in 51 of 133 patients [63.3%] vs 274 of 983 patients [27.9%]; P < .001). Finally,
(38.3%) versus 242 of 880 patients (27.5%), respectively in patients who had MDS with aberrations of chromosome
(P ¼ .014). 7 (vs patients who had MDS without aberrations of chromo-
Among the patients who had RAEB with 10% to some 7), CD56 expression was observed in 10 of 14 patients
19% excess blasts (RAEB-2) (vs patients without RAEB- (71.4%) versus 283 of 999 patients (28.3%; P ¼ .001).
2), CD13 negativity was observed in 11 of 81 patients
(13.6%) versus 52 of 932 patients (5.6%) (P ¼ .013); Aberrant antigen expression in erythrocytes
CD56 expression was observed in 41 of 81 patients Aberrant CD71 expression was observed signifi-
(50.6%) versus 252 of 932 (27%; P < .001); and CD2 cantly more often in both in patients who had MDS and
expression was observed in 12 of 81 patients (14.8%) ver- in patients who had suspected MDS (judged by CM)
sus 40 of 932 patients (4.3%; P < .001). compared with patients who did not have MDS (Table
In patients with RA (vs patients without RA), CD56 9). In patients who had RA, RARS, and RCMD/ringed
expression in monocytes was observed more often (17 of sideroblasts, compared with the respective remaining
31 patients [54.8%] vs 276 of 928 patients [28.1%]; patients who did not have those findings, homogeneously
P ¼ .002). In both patients with RA and patients with strong expression of CD71 was observed in 5 of 31
RARS, at least 2 aberrantly expressed antigens in mono- patients (16.1%) versus 62 of 982 patients (6.3%;
cytes were observed in 6 of 31 patients (19.4%) and in 2 P ¼ .048), in 6 of 27 patients (22.2%) versus 61 of 986
of 27 patients (7.4%), respectively. patients (6.2%; P ¼ .007), and in 8 of 49 patients

4558 Cancer October 1, 2010


MFC in MDS/Kern et al

Table 9. Correlation of Aberrant Antigen Expression in Erythrocytes With Cytomorphologya

Cytomorphologic Findings: No. of


Patients (%)
MFC Findings No MDS, MDS, Suspected P
n5277 n5511 MDS, n5225
CD71 homogeneously strong 3 (1.1) 46 (9) 18 (8) <.001
CD71 negative 6 (2.2) 71 (13.9) 17 (7.6) <.001

No. of aberrant antigens 0.00.2


per patient, meanSD
1 0.30.4 <.001
2 0.20.4 <.001

MFC indicates multiparameter flow cytometry; MDS, myelodysplastic syndrome; CD, cluster of differentiation; CD71,
transferrin receptor 1; SD, standard deviation.
a
Numbers of patients were compared using chi-square tests, and mean values were determined using Student t tests.

(16.3%) versus 59 of 964 patients (6.1%; (P ¼ .012),


respectively. In patients with RAEB-1 and RAEB-2, com-
pared with the respective remaining patients without
those findings, CD71 negativity was observed in 21 of
133 patients (15.8%) versus 73 of 880 patients (8.3%;
P ¼ .009) and in 14 of 81 patients (17.3%) versus 80 of
932 patients (8.6%; (P ¼ .015), respectively.
In the presence of CG abnormalities (vs normal CG
findings), greater frequencies of homogeneously strong
CD71 expression (27 of 245 patients [11%] vs 40 of 768
patients [5.2%]; P ¼ .003) and of CD71 negativity (32 of
245 patients [13.1%] vs 62 of 768 patients [8.1%];
P ¼ .023) were observed. In particular, in patients who had
5q as a sole CG abnormality, CD71 negativity was pres-
ent in 9 of 43 patients (20.9%) versus 85 of 970 patients
without 5q as a sole abnormality (8.8%; P ¼ .014).

Correlation of the Total Numbers of


Aberrantly Expressed Antigens With
Cytomorphology
The median total numbers of aberrantly expressed anti-
gens in blasts, granulocytes, monocytes, and erythrocytes
were 3 (range, 0-11), 1 (range, 0-8), and 0 (range, 0-7) in
patients from each group with CM results of MDS, sus-
pected MDS, and no MDS, respectively (P < .001) (Fig.
6). Using the CM result as the diagnostic gold standard,
different total numbers of aberrantly expressed antigens
were analyzed with regard to the sensitivity and specificity
of MFC in reproducing CM results. Table 10 demon-
strates that, although specificity remains at similar levels Figure 6. The total numbers of aberrant antigen expression in
blasts, granulocytes, monocytes, and erythrocytes are illus-
when applying higher numbers of aberrantly expressed trated for patients who were diagnosed by cytomorphology
antigens, a gradual loss of sensitivity is observed. with (A) no myelodysplastic syndrome, (B) myelodysplastic
syndrome, or (C) suspected myelodysplastic syndrome. The
A cutoff value of 6.3 for the SSC granulocyte:lym- height of each column indicates the numbers of patients.
phocyte ratio was identified as the best discriminate

Cancer October 1, 2010 4559


Original Article

Table 10. Sensitivity and Specificity of Multiparameter Flow Cytometry in Reproducing Cytomorphologic Diagnostic Results

Aberrantly Expressed Aberrantly Expressed Aberrantly Expressed


Antigens Only, % Antigens or Blast Antigens or Blast
Count > 5%, % Count > 5% or SSC
Ratio (Granulocytes:
Lymphocytes) > 6.3, %
MFC Criteria: Sensitivity Specificity Sensitivity Specificity Sensitivity Specificity
No. of Aberrantly
Expressed Antigens

2 75.5 92.6 79.6 91.3 85.3 86.3


3 57.5 93.6 65.2 92 77.3 86.6
4 32.9 95.5 46.8 92.6 66.7 85.9
5 16.9 94.5 35 91.3 61.6 85.4

SSC indicates side scatter; MFC, multiparameter flow cytometry.

Table 11. Relation Between the Total Number of Aberrantly


Expressed Antigens in All Cell Lineages and International
Prognostic Scoring System Scorea

IPSS No. of No. of Aberrantly


Score Patients Expressed Antigens,
Mean6SD
0 99 2.261.65
0.5 123 2.781.93
1.0 85 2.991.82
1.5 47 3.191.39
2.0 56 3.341.81
2.5 14 4.002.08
3.0 7 2.431.27

IPPS indicates International Prognostic Scoring System; SD, standard


deviation.
a
Data on IPSS scores were evaluable for 431 patients.

Figure 7. This Kaplan-Meier plot illustrates overall survival for


between results that were rated as MDS and no MDS by patients who had any of the 3 parameters 1) 3 aberrantly
CM. Taking into consideration an SSC granulocyte:lym- expressed antigens, 2) a blast count >5%, or 3) a side scatter
phocyte ratio of >6.3 and a blast count by MFC of >5% ratio (granulocytes:lymphocytes) >6.3 (solid line) versus
patients who had none of those parameters (dotted line;
and combining these parameters with the total number of 10-year over survival rate, 68% vs 100%, respectively; P ¼.008).
aberrantly expressed antigens resulted in improved sensi-
tivity (Table 10).
lyzed. OS, as determined from the date of a diagnosis or a
Relation of Aberrant Antigen Expression to suspected diagnosis of MDS to death, amounted to 74%
the International Prognostic Scoring System at 6 years. The number of aberrantly expressed antigens
The relation of MFC results to IPSS was assessed in 431 had a trend toward a correlation with OS (relative risk,
patients with MDS who had IPSS scores available. Table 1.2; P ¼ .09). Three parameters that were related to OS in
11 demonstrates that the total number of aberrantly univariate analyses were combined for an overall analysis
expressed antigens gradually increases with increasing of the relation between MDS-related findings by MFC
IPSS score (Spearman: r ¼ 0.409; P < .001). and OS: Those parameters were 3 aberrantly expressed
antigens, a blast count >5% in MFC, and an SSC granu-
Relation of Aberrant Antigen Expression locyte:lymphocyte ratio >6.3. Separating patients accord-
to Overall Survival ing to the presence of either versus none of these
In 257 patients who had clinical follow-up, the relation parameters resulted in significant differences in the 2-year
between MFC findings and overall survival (OS) was ana- OS rate (86% vs 100%; P ¼ .008) (Fig. 7).

4560 Cancer October 1, 2010


MFC in MDS/Kern et al

DISCUSSION rule out of MDS. Thus, the current series represents a per-
Standard BM evaluation in suspected MDS includes CM, fect cohort for the assessment of the potential of MFC in
cytochemistry, and iron staining as well as a CG analysis for the diagnostic workup of patients with unclear cytopenias
classification according to WHO8 and estimation of prog- and a differential diagnosis of MDS. For the first time to
nosis.10,11 However, many patients who have inconclusive our knowledge, the current study allows an evaluation of
CM results and a normal karyotype remain9,32,33 for the diagnostic usefulness of MFC in MDS in the context
whom an MDS cannot be diagnosed or ruled out. MFC is of a large cohort of patients characterized by CM and CG.
capable of identifying dysplastic features in BM samples In patients who had unequivocal CM findings, we
and, thus, was supposed to be added to the diagnostic observed a high degree of concordance (82%) between
workup in patients who have suspected MDS.13-16,32,34 CM and MFC results. It is noteworthy that, in patients
In the current study, several aspects regarding the who had dysplastic features that were insufficient to diag-
specificity and sensitivity of MFC for diagnosing MDS nose MDS using CM alone, aberrant antigen expression
have been elucidated in agreement with previous stud- was observed with MFC, although at lower frequencies
ies.15,16 In the current series and in the data reported by compared with the frequencies observed in patients with
van de Loosdrecht et al,16 this also is true for the identifi- clear-cut MDS. Furthermore, 13 of 277 patients (4.7%)
cation of dysplastic features in cell lineages that were not who had no evidence of MDS by CM were considered in
rated dysplastic by CM alone. agreement with MDS by MFC, posing the question of the
The enumeration of BM blasts has major prognostic specificity of MFC findings or the diagnostic role of MFC
impact10 and also guides the WHO classification.8 We in addition to CM. A definite proof of MDS could be
observed a significant degree of correlation and concord- accomplished in 6 such patients in the current series based
ance; however, patients who had counts that differed on MDS-typical CG aberrations, arguing in favor of the
between both methods also were encountered. Although a significance of MFC findings (which have been debated
difference in sample quality always may be considered as a during the revision of the WHO classification).9
cause, methodologic aspects also have to be considered. Thus, the current data suggest that, in patients who
First, smears for CM investigation and BM samples after have normal karyotypes, MFC also may identify individu-
Ficoll processing for MFC, per se, are different sources. als with MDS who have CM results that indicate either
Second, it is suggested that myeloid blasts should be iden- no evidence of MDS or dysplastic characteristics that are
tified in MFC by using multiple parameters, including not sufficient to clearly diagnose MDS. Consequently,
CD45/SSC signal and the expression of HLA-DR and although the diagnosis of MDS is based on CM results
CD34, together with myeloid markers.35 It has to be according to the WHO classification,8 it is noteworthy
taken into consideration, however, that cells counted as that CM per se does not have 100% sensitivity; therefore,
myeloid blasts by CM may fall into the monocyte cate- an analysis of the sensitivity and specificity of MFC in
gory in MFC and, thus, may be identified as dysplastic diagnosing MDS using CM as the gold standard is ham-
monocytes, which are not easily identified by CM and pered, and the respective results should be interpreted
need at least nonspecific-esterase evaluation. Along this accordingly. This was proven in the current analysis at
line, we observed that higher percentages of blasts were least in some of the patients who had positive MFC
determined by CM in samples that had aberrant antigen results, negative CM results, and CG results that revealed
expression in monocytes. Therefore, despite the high MDS-typical chromosomal aberrations. This scenario
degree of concordance, the percentages of blasts deter- further supports the need for clinical validation of the
mined by CM and by MFC should be considered sepa- diagnostic use of MFC in MDS, which should be eval-
rately. Consequently, the prognostic value of blast counts uated together with patient outcomes.36
determined by flow cytometry should be evaluated in pro- It is also clear, however, that, in the current series,
spective multicenter studies. 129 patients who had MDS clearly proven by CM did not
Although the majority of previous reports focused have dysplastic features identified by MFC. Thus, the
on patients with proven MDS and, in part, added control results of this crude analysis are suggestive of a combined
samples from patients who clearly did not have a malig- use of CM and MFC in the diagnostic evaluation of
nancy,13-16 in the current study, a large series of 1013 patients with suspected MDS.
patients with suspected MDS was the focus of our analy- Although we were able to reproduce some previously
ses. None of these patients had undergone a prior proof or reported findings, like the aberrant CD56 expression in

Cancer October 1, 2010 4561


Original Article

monocytes in CMML,37 the current study adds important MFC in a combined approach to optimize the overall
data on the capability of MFC to identify cell compart- diagnostic yield. Thus, the current gold standard for diag-
ments affected by dysplasia that are not identified by CM. nosing MDS, ie, CM, should be used as such but with the
This was reported previously by van de Loosdrecht et al16 awareness that other methods (ie, CG and MFC) may
and was reproduced here by the identification of an aber- identify patients with MDS who have results that are not
rant antigen expression in granulocytes in patients who rated MDS by CM. Accordingly, as the most reasonable
had no dysgranulopoiesis according to the CM results. approach to diagnosing MDS, we suggest a combination
One important aspect of handling data generated by of CM, CG, and MFC. Further studies should be per-
MFC in MDS is the definition of criteria that are useful formed to confirm these results and especially to define
to define MDS by MFC. Wells et al demonstrated that standards for the application of MFC in the diagnostic
the diagnostic specificity of MFC increases together with workup of MDS.34,35
an increase in the number of aberrant markers detected,
although at the cost of decreasing sensitivity.15 We made a
similar observation, although a specificity of 100% could CONFLICT OF INTEREST DISCLOSURES
not be reached in our dataset. This difference may be The authors made no disclosures.
because, in the current series, some patients without evi-
dence of MDS in CM analysis had MDS proven in CG
analysis, as discussed above. Furthermore, it has to be REFERENCES
stressed that the context of the current analysis is unique, 1. Cazzola M, Malcovati L. Myelodysplastic syndromes—cop-
in that a large number of patients with yet unclear cytope- ing with ineffective hematopoiesis. N Engl J Med. 2005;352:
536-538.
nias and with a differential diagnosis of MDS was assessed 2. Sole F, Luno E, Sanzo C, et al. Identification of novel cyto-
in parallel by the 2 gold-standard diagnostic methods, ie, genetic markers with prognostic significance in a series of
CM and CG, complemented by MFC. Thus, we were 968 patients with primary myelodysplastic syndromes. Hae-
matologica. 2005;90:1168-1178.
able to analyze both the specificity and the sensitivity of 3. Haase D. Cytogenetic features in myelodysplastic syn-
MFC findings in a routine diagnostic setting. These dromes. Ann Hematol. 2008;87:515-526.
results are in line with a previous report on the diagnostic 4. Fenaux P. Chromosome and molecular abnormalities in mye-
value of MFC in MDS that was based on a smaller series lodysplastic syndromes. Int J Hematol. 2001;73:429-437.
5. Bennett JM, Catovsky D, Daniel MT, et al. Proposals for
of 124 retrospectively analyzed patients in which both the classification of the myelodysplastic syndromes. Br J
CM and CG were used to validate MFC.38 Haematol. 1982;51:189-199.
In the current series, a correlation between an 6. Goasguen JE, Matsuo T, Cox C, Bennett JM. Evaluation of
the dysmyelopoiesis in 336 patients with de novo acute my-
increasing number of aberrantly expressed antigens and eloid leukemia: major importance of dysgranulopoiesis for
an increasing IPSS score was observed, and this finding remission and survival. Leukemia. 1992;6:520-525.
confirmed previous reports in smaller series.16 In addi- 7. Jaffe ES, Harris NL, Stein H, Vardiman JW, eds. World
Health Organization Classification of Tumours. Pathology
tion, in a subset of 257 patients, OS was inferior if MDS- and Genetics of Tumours of Haematopoietic and Lymphoid
typical findings by MFC were present. This also is in line Tissues. Lyon, France: IARC Press; 2001.
with previous data on patients with MDS who underwent 8. Jaffe ES, Harris NL, Stein H, Vardiman JW, eds. World
stem cell transplantation.15 Health Organization Classification of Tumours: Pathology
and Genetics of Tumours of Haematopoietic and Lymphoid
In conclusion, the current results demonstrate that, Tissues. Lyon, France: IARC Press; 2008.
in a large series of patients who had a differential diagnosis 9. Vardiman JW, Thiele J, Arber DA, et al. The 2008 revision
of MDS, MFC in combination with CM and CG may of the WHO classification of myeloid neoplasms and acute
leukemia: rationale and important changes. Blood. 2009;
add important diagnostic information. Patients who have 114:937-951.
aberrant antigen expression can be identified although 10. Greenberg P, Cox C, LeBeau M, et al. International scoring
their CM results may indicate no features of MDS. Cell system for evaluating prognosis in myelodysplastic syn-
dromes. Blood. 1997;89:2079-2088.
compartments that are affected by dysplasia and are not 11. Malcovati L, Germing U, Kuendgen A, et al. Time-depend-
identified by CM can be identified by MFC criteria. The ent prognostic scoring system for predicting survival and
further comparison based on CG and clinical data leukemic evolution in myelodysplastic syndromes. J Clin
strengthens the significance of the current MFC findings. Oncol. 2007;25:3503-3510.
12. Howe RB, Porwit-MacDonald A, Wanat R, Tehranchi R,
Because the sensitivity of both CM and MFC for identify- Hellstrom-Lindberg E. The WHO classification of MDS
ing MDS is <100%, we suggest considering CM and does make a difference. Blood. 2004;103:3265-3270.

4562 Cancer October 1, 2010


MFC in MDS/Kern et al

13. Ogata K, Nakamura K, Yokose N, et al. Clinical signifi- 25. Schoch C, Haferlach T, Haase D, et al. Patients with de
cance of phenotypic features of blasts in patients with mye- novo acute myeloid leukaemia and complex karyotype aber-
lodysplastic syndrome. Blood. 2002;100:3887-3896. rations show a poor prognosis despite intensive treatment: a
14. Stetler-Stevenson M, Arthur DC, Jabbour N, et al. Diagnos- study of 90 patients. Br J Haematol. 2001;112:118-126.
tic utility of flow cytometric immunophenotyping in myelo- 26. Schoch C, Kern W, Kohlmann A, Hiddemann W, Schnittger
dysplastic syndrome. Blood. 2001;98:979-987. S, Haferlach T. Acute myeloid leukemia with a complex
15. Wells DA, Benesch M, Loken MR, et al. Myeloid and aberrant karyotype is a distinct biological entity characterized
monocytic dyspoiesis as determined by flow cytometric scor- by genomic imbalances and a specific gene expression profile.
ing in myelodysplastic syndrome correlates with the IPSS Genes Chromosomes Cancer. 2005;43:227-238.
and with outcome after hematopoietic stem cell transplanta- 27. Haferlach C, Rieder H, Lillington DM, et al. Proposals for
tion. Blood. 2003;102:394-403. standardized protocols for cytogenetic analyses of acute leu-
16. van de Loosdrecht AA, Westers TM, Westra AH, Drager kemias, chronic lymphocytic leukemia, chronic myeloid leuke-
AM, van der Velden VH, Ossenkoppele GJ. Identification mia, chronic myeloproliferative disorders, and myelodysplastic
of distinct prognostic subgroups in low- and intermediate-1- syndromes. Genes Chromosomes Cancer. 2007;46:494-499.
risk myelodysplastic syndromes by flow cytometry. Blood. 28. Orfao A, Ortuno F, de Santiago M, Lopez A, San Miguel J.
2008;111:1067-1077. Immunophenotyping of acute leukemias and myelodysplas-
17. Haferlach T, Schoch C, Loffler H, et al. Morphologic dys- tic syndromes. Cytometry. 2004;58A:62-71.
plasia in de novo acute myeloid leukemia (AML) is related 29. Karmon Y, Manaster J, Chezar J. Immunophenotypic char-
to unfavorable cytogenetics but has no independent prog- acterization of myelopoiesis in early and late myelodysplastic
nostic relevance under the conditions of intensive induction syndromes: use of CD44 as an aid in early diagnosis.
therapy: results of a multiparameter analysis from the Ger- Cytometry. 2002;50:225-230.
man AML Cooperative Group studies. J Clin Oncol. 2003; 30. Borowitz MJ, Guenther KL, Shults KE, Stelzer GT. Immuno-
21:256-265. phenotyping of acute leukemia by flow cytometric analysis.
18. Haferlach T, Winkemann M, Loffler H, et al. The abnor- Use of CD45 and right-angle light scatter to gate on leukemic
mal eosinophils are part of the leukemic cell population in blasts in 3-color analysis. Am J Clin Pathol. 1993;100:534-540.
acute myelomonocytic leukemia with abnormal eosinophils 31. Kaplan EL, Meier P. Nonparametric estimation from
(AML M4Eo) and carry the pericentric inversion 16: a com- incomplete observations. J Am Stat Assoc. 1958;53:457-481.
bination of May-Grunwald-Giemsa staining and fluores- 32. Valent P, Horny HP, Bennett JM, et al. Definitions and
cence in situ hybridization. Blood. 1996;87:2459-2463. standards in the diagnosis and treatment of the myelodys-
19. Haferlach T, Kohlmann A, Schnittger S, et al. AML M3 plastic syndromes: consensus statements and report from a
and AML M3 variant each have a distinct gene expression working conference. Leuk Res. 2007;31:727-736.
signature but also share patterns different from other geneti- 33. Valent P, Hofmann WK, Busche G, et al. Meeting report:
cally defined AML subtypes. Genes Chromosomes Cancer. Vienna 2008 Workshop of the German-Austrian Working
2005;43:113-127. Group for Studying Prognostic Factors in Myelodysplastic
20. Bennett JM, Catovsky D, Daniel MT, et al. Proposal for Syndromes. Ann Hematol. 2009;88:607-611.
the recognition of minimally differentiated acute myeloid 34. van de Loosdrecht AA, Alhan C, Bene MC, et al. Standardi-
leukaemia (AML-MO) [see comments]. Br J Haematol. 1991; zation of flow cytometry in myelodysplastic syndromes:
78:325-329. report from the first European LeukemiaNet working con-
21. Bennett JM, Catovsky D, Daniel MT, et al. Proposed re- ference on flow cytometry in myelodysplastic syndromes.
vised criteria for the classification of acute myeloid leuke- Haematologica. 2009;94:1124-1134.
mia. A report of the French-American-British Cooperative 35. Loken MR, van de LA, Ogata K, Orfao A, Wells DA. Flow
Group. Ann Intern Med. 1985;103:620-625. cytometry in myelodysplastic syndromes: report from a
22. Bennett JM, Catovsky D, Daniel MT, et al. Proposals for working conference. Leuk Res. 2008;32:5-17.
the classification of the acute leukaemias. French-American- 36. Truong F, Smith BR, Stachurski D, et al. The utility of
British (FAB) Cooperative Group. Br J Haematol. 1976;33: flow cytometric immunophenotyping in cytopenic patients
451-458. with a non-diagnostic bone marrow: a prospective study.
23. Schoch C, Haferlach T, Bursch S, et al. Loss of genetic ma- Leuk Res. 2009;33:1039-1046.
terial is more common than gain in acute myeloid leukemia 37. Xu Y, McKenna RW, Karandikar NJ, Pildain AJ, Kroft SH.
with complex aberrant karyotype: a detailed analysis of 125 Flow cytometric analysis of monocytes as a tool for distin-
cases using conventional chromosome analysis and fluores- guishing chronic myelomonocytic leukemia from reactive
cence in situ hybridization including 24-color FISH. Genes monocytosis. Am J Clin Pathol. 2005;124:799-806.
Chromosomes Cancer. 2002;35:20-29. 38. Kussick SJ, Fromm JR, Rossini A, et al. Four-color flow
24. Schoch C, Kern W, Krawitz P, et al. Dependence of age- cytometry shows strong concordance with bone marrow
specific incidence of acute myeloid leukemia on karyotype. morphology and cytogenetics in the evaluation for myelo-
Blood. 2001;98:3500. dysplasia. Am J Clin Pathol. 2005;124:170-181.

Cancer October 1, 2010 4563

You might also like