Multiparameter flow cytometry immunophenotyping allows discrimination between normal (N-) and myelomatous (MM-) plasma cells (PCs) within the bone marrow plasma cell compartment (BMPCs). Here we report on the prognostic relevance of detecting more than 5% residual normal plasma cells from all bone marrow plasma cells (N-PCs/BMPCs) by multiparameter flow cytometry in a series of 594 newly diagnosed symptomatic MM patients, uniformly treated according to the Grupo Español de MM 2000 (GEM2000) protocol. Our results show that symptomatic MM patients with more than 5% N-PCs/BMPCs (n = 80 of 594; 14%) have a favorable baseline clinical prospect, together with a significantly lower frequency of high-risk cytogenetic abnormalities and higher response rates. Moreover, this group of patients had a significantly longer progression-free survival (median, 54 vs 42 months, P = .001) and overall survival (median, not reached vs 89 months, P = .04) than patients with less than or equal to 5% N-PCs/BMPCs. Our findings support the clinical value of detecting residual normal PCs in MM patients at diagnosis because this reveals a good prognostic category that could benefit from specific therapeutic approaches. This trial was registered at www.clinicaltrials.gov as NCT00560053.

Multiparameter flow cytometry (MFC) immunophenotyping allows discrimination between myelomatous plasma cells (MM-PCs) and normal/reactive PC (N-PCs).1-4  This has been used for the differential diagnosis between monoclonal gammopathy of undetermined significance (MGUS) and multiple myeloma (MM),3  the identification of high-risk MGUS and smoldering MM (SMM) patients,4  minimal residual disease investigation,5-8  definition of prognostic antigenic profiles,1  and identification of new therapeutic targets.2  Typically, in symptomatic MM patients at diagnosis, most bone marrow plasma cells (BMPCs) are clonal, and N-PCs from all BMPCs are only detected in a minority of MM cases.3,9  By contrast, 82% of MGUS and 40% of SMM patients show more than 5% residual normal plasma cells from all bone marrow plasma cells (N-PCs/BMPCs), in association with a low risk of progression to symptomatic MM.4  Here we report on the results of a prospective analysis of the prognostic impact of the presence of more than 5% N-PCs/BMPCs at diagnosis in a large series of uniformly treated symptomatic MM patients.

Patients

The study included 594 untreated, symptomatic MM patients diagnosed according to the International Myeloma Working Group criteria10  and uniformly treated following the Spanish GEM2000 protocol (VBMCP/VBAD ×6 plus autologous stem cell transplantation [ASCT]).1,6  Written informed consent was obtained in accordance with the Declaration of Helsinki, as well as Institutional Review Board approval from each participating hospital. Response was assessed using the European Group for Blood and Marrow Transplant criteria.11,12  At the study endpoint, 380 patients (64%) relapsed/progressed and 188 (32%) died, with a median follow-up of 54 months; median progression-free survival (PFS) and overall survival (OS) were 44 and 97 months, respectively. It is the current standard of clinical practice in MM to report cytogenetic results on purified PCs,13  but unfortunately this was not part of the initial workload of the GEM2000 protocol. Therefore, we used a complementary series of 501 patients with complete cytogenetic/fluorescence in situ hybridization data from immunomagnetic-enriched plasma cells, with a rather short follow-up, which precludes survival analysis. Information of serum-free light chain (sFLC) was also extracted from this series.

MFC immunophenotypic studies

Erythrocyte-lysed whole BM samples were stained using a 4-color direct immunofluorescence technique, previously described in detail.1,6,8  The following monoclonal antibody combinations (fluorescein isothiocyanate/phycoerythrin/peridinin chlorophyll protein-Cy5.5/allophycocyanin) were used to identify aberrant antigen expression in PCs: CD38/CD56/CD19/CD45, CD138/CD28/CD33/CD38, and CD20/CD117/CD138/CD38.1,6,8,14  Acquisition was performed in a FACSCalibur flow cytometer (BD Biosciences) using the CellQuest program (BD Biosciences).1,2,4,6,8  Information was recorded for at least 3 × 103 BMPCs/tube. The Paint-A-Gate PRO program (BD Biosciences) was used for data analysis, following the recommendations of the European Myeloma Network.14 

Statistical analyses

The χ2 and Mann-Whitney U tests were used to estimate statistical significant differences. Survival curves were plotted using the Kaplan-Meier method, assessing differences with the log-rank test. For the multivariate analysis, the Cox regression proportional hazard model (stepwise regression) was used. Statistical analyses were carried out with SPSS (Version 15.0; SPSS Inc).

Two groups have identified the presence on N-PCs in MM patients at diagnosis in a small series.9,15  However, neither the number of these cells nor the clinical and prognostic value was analyzed. Here, 80 of 594 newly diagnosed MM patients (14%) had more than 5% N-PCs/BMPCs by MFC, all of them with CRAB features (increased calcium, renal insufficiency, anaemia, or bone lesions). Interestingly, the presence of more than 5% N-PCs/BMPCs was associated with characteristics related to a favorable prognosis16-19  (Table 1): lower β2 microglobulin (β)2M (P = .01) and M-component serum levels (P < .001), lower BMPC infiltration by both morphology (P < .001) and MFC (P < .001), together with higher hemoglobin levels (P < .001), among other features (Table 1). In addition, immunoparesis (reduction below the lower normal limit in the levels of one or 2 uninvolved Ig) was significantly less frequent among patients with more than 5% N-PCs/BMPCs than in those with less than or equal to 5% N-PCs/BMPCs (42% vs 83%, respectively; P = .003), probably reflecting higher levels of residual N-PCs. Concerning the MM-PC immunophenotypic profile, the only antigen showing significant differences was CD117, a phenotypic marker associated with a favorable outcome,1,20,21  with cases holding more than 5% N-PCs/BMPCs showing a higher incidence of positive CD117 expression (49% vs 34%, respectively; P = .04). In the complementary series of patients with complete cytogenetic/fluorescence in situ hybridization information (Table 2), the frequency of cases with more than 5% N-PCs/BMPCs (12%; Table 1) was almost identical, which excludes a bias in patient selection. Interestingly, patients with more than 5% N-PCs/BMPCs had a lower incidence of del (13q) (5% vs 41%; P < .001) and IgH translocations (13% vs 43%; P < .001) than cases with less than or equal to 5% N-PCs/BMPCs. Moreover, whereas the incidence of t(11;14), which has little influence on survival, was similar in both group of patients (13% vs 18%; not significant), t(4;14) was not detected in cases with more than 5% N-PCs/BMPCs, although it represented 15% of cases with less than or equal to 5% N-PCs/BMPCs (P = .002). In addition, when high-risk cytogenetic cases [t(4;14), t(4;16), or del (17p)] were grouped, we founded a significantly lower frequency in cases with more than 5% N-PCs/BMPCs (3% vs 26%, respectively; P = .006). Moreover, considering patients with sFLC data available (N = 261) at diagnosis, cases with more than 5% N-PCs/BMPCs presented a higher frequency of normal sFLC ratio than those with less than or equal to 5% N-PCs/BMPCs (10 of 33, 30%; vs 16 of 228, 7%; P < .001).

Table 1

Patient demographics and baseline characteristics of symptomatic MM patients

Patient demographics and baseline characteristics% of normal PC from all BMPC (N-PC/BMPC)
P
≤ 5% (n = 514)> 5% (n = 80)
Male/female* 283 (55%)/231 (45%) 48 (60%)/32 (40%) NS 
Age, y 58 (32-70) 58 (35-70) NS 
Subtype of MM*    
    IgG 304 (59%) 37 (46%) .06 
    IgA 123 (24%) 17 (22%) NS 
    Bence-Jones protein 82 (16%) 22 (27%) .04 
    Nonsecretory 5 (1%) 4 (5%) NS 
ISS*    
    Stage I 180 (35%) 38 (48%) .03 
    Stage II 226 (44%) 30 (37%) NS 
    Stage III 108 (21%) 12 (15%) NS 
β2-microglobulin, mg/L 3.2 (0.09-41) 2.5 (0-24) .009 
Hemoglobin, g/L 106 (10-167) 121 (60-168) < .001 
Albumin, g/dL 3.6 (0.3-7) 3.8 (1.8-6.4) .02 
Platelet count, ×109/L 207 (6-990) 241 (58-486) < .001 
M-component, g/dL 4 (0-47) 2 (0-22) < .001 
Immunoparesis, 1 or 2 Ig* 427 (83%) 34 (42%) .003 
Plasma cells by morphology, % 40% (1-100%) 17 (1-95) < .001 
Plasma cells by MFC, % 13 (0.1-90) 1.6 (0.05-22) < .001 
Plasma cells in S-phase, % 1.4 (0-15) 2.0 (0-15) NS 
CD117+ cases by MFC, n (%)* 175 (34) 39 (49) .04 
Patient demographics and baseline characteristics% of normal PC from all BMPC (N-PC/BMPC)
P
≤ 5% (n = 514)> 5% (n = 80)
Male/female* 283 (55%)/231 (45%) 48 (60%)/32 (40%) NS 
Age, y 58 (32-70) 58 (35-70) NS 
Subtype of MM*    
    IgG 304 (59%) 37 (46%) .06 
    IgA 123 (24%) 17 (22%) NS 
    Bence-Jones protein 82 (16%) 22 (27%) .04 
    Nonsecretory 5 (1%) 4 (5%) NS 
ISS*    
    Stage I 180 (35%) 38 (48%) .03 
    Stage II 226 (44%) 30 (37%) NS 
    Stage III 108 (21%) 12 (15%) NS 
β2-microglobulin, mg/L 3.2 (0.09-41) 2.5 (0-24) .009 
Hemoglobin, g/L 106 (10-167) 121 (60-168) < .001 
Albumin, g/dL 3.6 (0.3-7) 3.8 (1.8-6.4) .02 
Platelet count, ×109/L 207 (6-990) 241 (58-486) < .001 
M-component, g/dL 4 (0-47) 2 (0-22) < .001 
Immunoparesis, 1 or 2 Ig* 427 (83%) 34 (42%) .003 
Plasma cells by morphology, % 40% (1-100%) 17 (1-95) < .001 
Plasma cells by MFC, % 13 (0.1-90) 1.6 (0.05-22) < .001 
Plasma cells in S-phase, % 1.4 (0-15) 2.0 (0-15) NS 
CD117+ cases by MFC, n (%)* 175 (34) 39 (49) .04 

Patient demographics and baseline characteristics of symptomatic MM patients grouped according to the percentage of N-PC/BMPC.

MM indicates multiple myeloma; N-PC/BMPC, normal plasma cells from all bone marrow plasma cells; PC, plasma cells; BMPC, bone marrow plasma cells; NS, statistically not significant (P > .05); ISS, international staging system; and MFC, multiparameter flow cytometry.

*

Results expressed as percentage of cases.

Results expressed as median (range).

Table 2

Cytogenetics

Cytogenetics% of normal PC from all BMPC (N-PC/BMPC)
P
≤ 5% (n = 439)> 5% (n = 62)
IgH translocations 187 (43%) 8 (13%) < .001 
    t(4;14) 66 (15%) 0 (0%) .002 
    t(11;14) 79 (18%) 8 (13%) NS 
    t(14;16) 18 (4%) 0 (0%) NS 
    Others 35 (8%) 0 (0%) .009 
Del (13q) 180 (41%) 3 (5%) < .001 
Del (17p) 36 (8%) 2 (3%) NS 
High-risk: any t(4;14), t(14;16), or del(17p)− 114 (26%) 2 (3%) .006 
Cytogenetics% of normal PC from all BMPC (N-PC/BMPC)
P
≤ 5% (n = 439)> 5% (n = 62)
IgH translocations 187 (43%) 8 (13%) < .001 
    t(4;14) 66 (15%) 0 (0%) .002 
    t(11;14) 79 (18%) 8 (13%) NS 
    t(14;16) 18 (4%) 0 (0%) NS 
    Others 35 (8%) 0 (0%) .009 
Del (13q) 180 (41%) 3 (5%) < .001 
Del (17p) 36 (8%) 2 (3%) NS 
High-risk: any t(4;14), t(14;16), or del(17p)− 114 (26%) 2 (3%) .006 

Information on patient cytogenetics corresponds to a parallel series of not uniformly treated MM patients, not to the GEM2000 series reported in the present paper.

Results expressed as percentage of cases.

NS indicates not statistically significant.

Concerning response to therapy (Table 3), patients with more than 5% compared with those with less than or equal to 5% N-PCs/BMPCs showed higher rates of complete remission after induction (21% vs 11%, respectively; P < .001), and at day 100 after ASCT (64% vs 33%, respectively; P < .001). Accordingly, cases with more than 5% N-PCs/BMPCs were more frequently minimal residual disease negative at day 100 after ASCT (68% vs 37%; P < .001) and also showed a faster and more complete recovery of the polyclonal PC population at day 100 after ASCT, compared with less than or equal to 5% N-PCs/BMPCs cases (mean ratio of N-PCs/BMPCs of 87 vs 64%, respectively; P < .001). Differences in response rates translated into distinct survival, as cases with more than 5% N-PCs/BMPCs showed a better outcome than patients with less than or equal to 5% N-PCs/BMPCs with significant differences in both PFS (median, 54 vs 42 months; P = .001) and OS (median, not reached vs 89 months, P = .04) (Figure 1). Therefore, the presence of more than 5% N-PCs/BMPCs emerges as a new prognostic factor for symptomatic MM patients, similar to what has previously been found in both MGUS and SMM.4  Other baseline significant factors for survival in the univariate analysis were: low hemoglobin (≤ 100 g/L; P ≤ .008), higher calcium (> 10 mg/dL; P ≤ .04), increased creatinine (> 2 mg/dL; P = .002) levels, higher percentages of BMPCs by microscopy (> 30% BMPCs; P ≤ .006) and MFC (> 15%BMPCs; P ≤ .007), advanced disease (International Staging System [ISS] stage III; P ≤ .004), and high-risk cytogenetics (P < .001). Despite the association found between the presence of more than 5% N-PCs/BMPCs and immunoparesis, it should be emphasized that the latter variable did not appear to have a significant impact on survival, suggesting that it cannot be used as a surrogate marker to replace immunophenotyping. Multivariate analysis performed in cases with cytogenetic information (N = 176) showed only cytogenetics as an independent prognostic factor for PFS (P < .001) and OS (P = .002). Because cytogenetic information was only available for a subset of patients, a new multivariate analysis was performed for the whole series. With respect to PFS, both the ISS disease stage (relative risk of progression [RR] = 1.4; P = .03) and the percentage of N-PCs/BMPCs (RR = 1.6; P = .008) were selected as independent prognostic factors, whereas for OS hemoglobin levels (RR = 1.6; P = .01) together with the ISS disease stage (RR = 2.1; P = .003) fitted the model.

Table 3

Response to therapy

EBMT criteria≤ 5% N = PC/BMPC
> 5% N = PC/BMPC
P
After inductionAfter ASCTAfter inductionAfter ASCTAfter inductionAfter ASCT
CR 56 (11%) 168 (33%) 17 (21%) 51 (64%) .01 < .001 
nCR 61 (12%) 99 (19%) 19 (24%) 8 (10%) .005 < .001 
PR or less 397 (77%) 247 (48%) 44 (55%) 21 (26%) < .001 < .001 
EBMT criteria≤ 5% N = PC/BMPC
> 5% N = PC/BMPC
P
After inductionAfter ASCTAfter inductionAfter ASCTAfter inductionAfter ASCT
CR 56 (11%) 168 (33%) 17 (21%) 51 (64%) .01 < .001 
nCR 61 (12%) 99 (19%) 19 (24%) 8 (10%) .005 < .001 
PR or less 397 (77%) 247 (48%) 44 (55%) 21 (26%) < .001 < .001 

Results are expressed as number (percentage) of cases. N = PC indicates normal plasma cells; BMPC, bone marrow plasma cells; and ASCT, autologus stem cell transplantation.

Figure 1

PFS and OS of symptomatic MM patients grouped according to the presence (N = 80) or absence (N = 514) of more than 5% N-PCs/BMPCs at diagnosis. (A) PFS. (B) OS.

Figure 1

PFS and OS of symptomatic MM patients grouped according to the presence (N = 80) or absence (N = 514) of more than 5% N-PCs/BMPCs at diagnosis. (A) PFS. (B) OS.

Close modal

In conclusion, our results show that in symptomatic MM patients the identification of a significant number of residual normal PCs at diagnosis (> 5% N-PCs/BMPCs) reveals a specific subgroup of patients with a unique biologic signature and prolonged survival who could benefit from specific therapeutic approaches.

The online version of this article contains a data supplement.

The publication costs of this article were defrayed in part by page charge payment. Therefore, and solely to indicate this fact, this article is hereby marked “advertisement” in accordance with 18 USC section 1734.

We thank all members of the GEM/PETHEMA group for their cooperation.

This work was supported by the Cooperative Research Thematic Network (RTICs; RD06/0020/0006), MM Jevitt, SL firm, and Instituto de Salud Carlos III/ Subdirección General de Investigación Sanitaria (FIS: PI060339, 02/0905, 01/0089/01-02, and PI060339).

Contribution: J.F.S.-M., J.J.L., and A.O. conceived the idea, and together with M.-B.V., designed the study protocol; B.P., M.-B.V., G.M., J.J.P., M.A.M., M.C.L.-B., and L.M. analyzed the flow cytometry data; A.S., N.C.G., A.G.d.C., N.d.l.H., M.V.M., R.G.-B., J.G., J.H., L.P., D.C., R.M., J.d.l.R., A.M., Y.G., J.J.L., and J.B. contributed with provision of study material or patients; B.P., M.-B.V., and G.M. collected and assembled data; B.P., M.-B.V., and J.F.S.-M. analyzed and interpreted data; B.P. and M.-B.V. performed statistical analysis; B.P., M.-B.V., and J.F.S.-M. wrote the manuscript; and all authors reviewed and approved the manuscript.

Conflict-of-interest disclosure: The authors declare no competing financial interests.

A complete list of GEM Cooperative Study Group participants appears in the online “Appendix” (available on the Blood website; see the Supplemental Materials link at the top of the online article).

Correspondence: Jesús F. San-Miguel, Hospital Universitario de Salamanca, Paseo de San Vicente 58-182, 37007 Salamanca, Spain; e-mail: sanmigiz@usal.es.

1
Mateo
 
G
Montalban
 
MA
Vidriales
 
MB
, et al. 
Prognostic value of immunophenotyping in multiple myeloma: a study by the PETHEMA/GEM cooperative study groups on patients uniformly treated with high-dose therapy.
J Clin Oncol
2008
, vol. 
26
 
16
(pg. 
2737
-
2744
)
2
Mateo
 
MG
San
 
M
I
 
Orfao de
 
MA
Immunophenotyping of plasma cells in multiple myeloma.
Methods Mol Med
2005
, vol. 
113
 (pg. 
5
-
24
)
3
Ocqueteau
 
M
Orfao
 
A
Almeida
 
J
, et al. 
Immunophenotypic characterization of plasma cells from monoclonal gammopathy of undetermined significance patients: implications for the differential diagnosis between MGUS and multiple myeloma.
Am J Pathol
1998
, vol. 
152
 
6
(pg. 
1655
-
1665
)
4
Perez-Persona
 
E
Vidriales
 
MB
Mateo
 
G
, et al. 
New criteria to identify risk of progression in monoclonal gammopathy of uncertain significance and smoldering multiple myeloma based on multiparameter flow cytometry analysis of bone marrow plasma cells.
Blood
2007
, vol. 
110
 
7
(pg. 
2586
-
2592
)
5
Liu
 
H
Yuan
 
C
Heinerich
 
J
, et al. 
Flow cytometric minimal residual disease monitoring in patients with multiple myeloma undergoing autologous stem cell transplantation: a retrospective study.
Leuk Lymphoma
2008
, vol. 
49
 
2
(pg. 
306
-
314
)
6
Paiva
 
B
Vidriales
 
MB
Cervero
 
J
, et al. 
Multiparameter flow cytometric remission is the most relevant prognostic factor for multiple myeloma patients who undergo autologous stem cell transplantation.
Blood
2008
, vol. 
112
 
10
(pg. 
4017
-
4023
)
7
Rawstron
 
AC
Davies
 
FE
Dasgupta
 
R
, et al. 
Flow cytometric disease monitoring in multiple myeloma: the relationship between normal and neoplastic plasma cells predicts outcome after transplantation.
Blood
2002
, vol. 
100
 
9
(pg. 
3095
-
4100
)
8
San Miguel
 
JF
Almeida
 
J
Mateo
 
G
, et al. 
Immunophenotypic evaluation of the plasma cell compartment in multiple myeloma: a tool for comparing the efficacy of different treatment strategies and predicting outcome.
Blood
2002
, vol. 
99
 
5
(pg. 
1853
-
1856
)
9
Olteanu
 
H
Wang
 
HY
Chen
 
W
McKenna
 
RW
Karandikar
 
NJ
Immunophenotypic studies of monoclonal gammopathy of undetermined significance.
BMC Clin Pathol
2008
, vol. 
8
 pg. 
13
 
10
International Myeloma Working Group
Criteria for the classification of monoclonal gammopathies, multiple myeloma and related disorders: a report of the International Myeloma Working Group.
Br J Haematol
2003
, vol. 
121
 
5
(pg. 
749
-
757
)
11
Blade
 
J
Samson
 
D
Reece
 
D
, et al. 
Criteria for evaluating disease response and progression in patients with multiple myeloma treated by high-dose therapy and haemopoietic stem cell transplantation: Myeloma Subcommittee of the EBMT. European Group for Blood and Marrow Transplant.
Br J Haematol
1998
, vol. 
102
 
5
(pg. 
1115
-
1123
)
12
Sarasquete
 
ME
Garcia-Sanz
 
R
Gonzalez
 
D
, et al. 
Minimal residual disease monitoring in multiple myeloma: a comparison between allelic-specific oligonucleotide real-time quantitative polymerase chain reaction and flow cytometry.
Haematologica
2005
, vol. 
90
 
10
(pg. 
1365
-
1372
)
13
Fonseca
 
R
Barlogie
 
B
Bataille
 
R
, et al. 
Genetics and cytogenetics of multiple myeloma: a workshop report.
Cancer Res
2004
, vol. 
64
 
4
(pg. 
1546
-
1558
)
14
Rawstron
 
AC
Orfao
 
A
Beksac
 
M
, et al. 
Report of the European Myeloma Network on multiparametric flow cytometry in multiple myeloma and related disorders.
Haematologica
2008
, vol. 
93
 
3
(pg. 
431
-
438
)
15
Sezer
 
O
Heider
 
U
Zavrski
 
I
Possinger
 
K
Differentiation of monoclonal gammopathy of undetermined significance and multiple myeloma using flow cytometric characteristics of plasma cells.
Haematologica
2001
, vol. 
86
 
8
(pg. 
837
-
843
)
16
Greipp
 
PR
Leong
 
T
Bennett
 
JM
, et al. 
Plasmablastic morphology: an independent prognostic factor with clinical and laboratory correlates. Eastern Cooperative Oncology Group (ECOG) myeloma trial E9486 report by the ECOG Myeloma Laboratory Group.
Blood
1998
, vol. 
91
 
7
(pg. 
2501
-
2507
)
17
San Miguel
 
JF
Sanchez
 
J
Gonzalez
 
M
Prognostic factors and classification in multiple myeloma.
Br J Cancer
1989
, vol. 
59
 
1
(pg. 
113
-
118
)
18
Greipp
 
PR
San
 
MJ
Durie
 
BG
, et al. 
International staging system for multiple myeloma.
J Clin Oncol
2005
, vol. 
23
 
15
(pg. 
3412
-
3420
)
19
Nowakowski
 
GS
Witzig
 
TE
Dingli
 
D
, et al. 
Circulating plasma cells detected by flow cytometry as a predictor of survival in 302 patients with newly diagnosed multiple myeloma.
Blood
2005
, vol. 
106
 
7
(pg. 
2276
-
2279
)
20
Mateo
 
G
Castellanos
 
M
Rasillo
 
A
, et al. 
Genetic abnormalities and patterns of antigenic expression in multiple myeloma.
Clin Cancer Res
2005
, vol. 
11
 
10
(pg. 
3661
-
3667
)
21
Bataille
 
R
Jego
 
G
Robillard
 
N
, et al. 
The phenotype of normal, reactive and malignant plasma cells: identification of “many and multiple myelomas” and of new targets for myeloma therapy.
Haematologica
2006
, vol. 
91
 
9
(pg. 
1234
-
1240
)

Author notes

*

B.P. and M.-B.V. contributed equally to this study.

Sign in via your Institution