TO THE EDITOR:

Induction and consolidation based on proteasome inhibitors, immunomodulatory drugs, and corticoids integrated with high-dose therapy (HDT) and autologous stem cell transplantation (ASCT), are showing complete response (CR) rates >50% in multiple myeloma (MM).1-3 The addition of anti-CD38 monoclonal antibodies may increase these unprecedented CR rates.4-6 When more than half of transplant-eligible patients with MM achieve CR with frontline therapy, it is reasonable to ask, what other tests are clinically relevant after negative immunofixation.

The achievement of deep responses with modern therapy led the International Myeloma Working Group (IMWG) to propose new guidelines that included definitions of negative minimal residual disease (MRD) for standard response criteria.7 Indeed, recent studies have reported nearly 50% MRD rates,5,8,9 and, more importantly, the prognostic value of MRD criteria was validated in clinical trials8,10-12 and routine practice.13,14 However, the clinical significance of standard response criteria in patients who are MRD positive has not been investigated in current treatment scenarios.

Four hundred forty-nine newly diagnosed, transplant-eligible patients enrolled in the phase 3 PETHEMA/GEM2012MENOS65 trial (registered on clinicaltrials.gov, as NCT01916252) and with available response assessment, were included in this study.1 Afterward, patients were enrolled in the PETHEMA/GEM2014MAIN trial (NCT02406144).8 Patients’ demographics and clinical features have been described elsewhere.1 Minimal Residual Disease (MRD) was assessed using next-generation flow (NGF) cytometry, as reported recently.8 An independent ethics committee approved the protocol, and informed consent forms were required before patients were enrolled. Studies were conducted per the ethical principles of the Declaration of Helsinki.

Response was assessed after the last induction cycle, at day 100 after HDT/ASCT, and after the second consolidation course. Seventy-four patients were not evaluated after consolidation. Response was defined per the 2016 guidelines7 with 2 exceptions: (1) patients with ≤5% bone marrow plasma cells (BMPCs) and negative serum immunofixation, but unavailable urine immunofixation data, were reclassified as attaining CR according to our recent findings indicating identical outcomes15; (2) patients showing <2 × 10−6 tumor cells were defined as having undetectable MRD, regardless of the depth of serological response, because the outcomes were identical between those with persistent and absent M-component.8 Differences were tested for statistical significance with the (2-sided) log-rank test, and hazards ratios (with its 2-sided 95% confidence interval) were estimated with a Cox regression model.

We started by analyzing the prognostic value of standard response criteria after 6 induction cycles with bortezomib, lenalidomide, and dexamethasone (VRD); HDT/ASCT; and consolidation with 2 VRD courses. With a median follow-up of 5 years, achieving CR or stringent CR (sCR) after induction resulted in significantly superior progression-free survival (PFS) when compared with very good partial response (VGPR) or PR (Figure 1A). After HDT/ASCT, patients in CR or sCR continued showing more prolonged PFS compared with those in VGPR, though not those in PR (Figure 1B). Surprisingly, the differences disappeared after consolidation (Figure 1C). Less than half of the patients in VGPR (39 of 86; 45%) and in PR (6 of 19; 32%) after consolidation have respectively achieved CR/sCR during the first year of maintenance, excluding deepening of response at later stages as a unifying reason to explain similar PFS between these patients and those in CR or sCR after consolidation. These data urge further investigations to understand the limitations of standard response criteria.

Figure 1.

Depth of response based on standard response criteria. PFS of patients achieving sCR, CR, VGPR, and PR after 6 induction cycles of VRD (A); ASCT conditioned with Bu-Mel or Mel-200 HDT (B); or 2 consolidation cycles of VRD (C). PFS was defined as time from response assessment until disease progression or death from any cause and was estimated by the Kaplan-Meier method. OS was defined as time from response assessment until death from any cause. *P < .05; **P < .01; ***P < .001.

Figure 1.

Depth of response based on standard response criteria. PFS of patients achieving sCR, CR, VGPR, and PR after 6 induction cycles of VRD (A); ASCT conditioned with Bu-Mel or Mel-200 HDT (B); or 2 consolidation cycles of VRD (C). PFS was defined as time from response assessment until disease progression or death from any cause and was estimated by the Kaplan-Meier method. OS was defined as time from response assessment until death from any cause. *P < .05; **P < .01; ***P < .001.

Close modal

In 1998, a cutoff of <5% BMPCs by conventional cytology was added into the CR criteria.16 Thus, we investigated whether it remains informative >20 years later. The median percentage of PCs by morphology was 1.7% (range, 0-5) in the 266 patients in CR or sCR after consolidation, and PC enumeration by morphology had no prognostic value (data not shown). Of note, only 4 of 270 (1.5%) patients with a negative immunofixation had >5% BMPCs and therefore, were not classified as being in CR at that time point (all 4 cases achieved CR later on; 1 had progressive disease and died, whereas the others remained progression free). This percentage is almost 10-fold inferior to that reported >10 years ago by Chee et al17 and stresses the limited value of cytological response assessment in transplant-eligible patients, with patients with MM reaching high-quality remission with novel drug combinations.

BM biopsies were first introduced in the 2006 response criteria18 to evaluate PC clonality using immunohistochemistry or immunofluorescence. Namely, it required the analysis of a minimum of 100 PCs and a κ/λ ratio of >4:1 or <1:2 in patients with MM with the κ or λ isotype. Accordingly, clonality was respectively defined whenever >80% total PCs stained for κ or >50% total PCs stained for λ. We found that the median percentage of clonal and normal PCs among total PCs identified by NGF in patients achieving CR or sCR after consolidation was 3% and 97%, respectively (supplemental Figure 1A). These findings uncover that the median percentage of normal PCs is 32-fold greater than that of clonal PCs within the PC compartment and expose that simple κ/λ ratios measured in 100 PCs7 do not detect such low levels of residual disease. Indeed, the median level of MRD was 0.03% (range, 0.0002% to 0.59%; supplemental Figure 1B) and only 1% of patients with κ and 3% of patients with λ MM, respectively, had >80% and >50% clonal PCs of the total PCs.

The serum free light-chain (sFLC) ratio was introduced alongside BM clonality, as previously defined, to create the sCR criteria.18 We found that almost one-fourth of patients in CR after consolidation display an abnormal sFLC ratio (72 of 266; 27%), but their PFS was identical with that of cases with a normal sFLC ratio (Figure 1C). Similar results were observed after induction and HDT/ASCT (Figure 1A-B). Altogether, these data raise questions about the sensitivity and clinical utility of immunohistochemistry and immunofluorescence and the sFLC assays in patients in CR treated with optimal intensive treatment. As a result, the superiority of sCR criteria over standard CR criteria could not be confirmed in this study or elsewhere.19 

We had shown with low-sensitivity flow cytometry that MRD was detected in approximately one-third of patients in CR who had been treated with older regimens.20-22 In this study, NGF enabled the detection of MRD in 73 of 252 (29%) patients in CR or sCR after consolidation and, in most cases, MRD levels ranged between 10−6 and 10−5 (58 of 73; 79%). These results endorse the IMWG 2016 response assessment guidelines that indicate a minimum sensitivity of 10−5,7 given that MRD criteria based on a threshold of 10−4 would have limited value in patients achieving high-quality remission induced by newer, more effective regimens. Accordingly, there were significant differences in PFS and OS (supplemental Figure 2) among patients in CR or sCR who had positive vs negative MRD, which further validates the inclusion of the former in the 2016 response criteria. Of note, transplant-eligible patients in CR or sCR with positive MRD after induction and consolidation with VRD showed a median PFS of 3 years. These findings further support that MRD negativity and not CR should be the new end point in transplant-eligible MM, although questions about the timing, periodicity, and sensitivity of MRD assessment remain unresolved.

Although the prognostic value of CR and sCR is limited when compared with that of MRD, it could be that standard response criteria are of prognostic value in patients who remain MRD+. However, we found no significant differences in PFS (P ≥ .089; Figure 2A) and OS (P ≥ .496; Figure 2B) across patients with persistent MRD, regardless of the achievement of sCR, CR, VGPR, or even PR. PFS rates at 5 years were 43%, 35%, 51%, and 40%, respectively; similar results were observed for OS (75%, 73.5%, 68%, and 66% at 5 years). These numbers were significantly inferior to those of patients achieving undetectable MRD (5-year PFS and OS of 79% and 93%, respectively). Thus, our results reproduce and expand previous observations of modern therapies, wherein attaining CR or sCR without MRD clearance is no better than a VGPR or PR in terms of PFS and OS.20 Of further note, patients in sCR, CR, or VGPR showed similar survival upon achieving undetectable MRD (supplemental Figure 3). Thus, and with the possible exception of patients with extramedullary disease and elevated LDH levels,8 undetectable MRD in cases of persistent M-component should not be generalized as a false-negative result.

Figure 2.

Depth of response based on standard and MRD response criteria. PFS (A) and OS (B) of patients stratified according to best response achieved during treatment: MRD, sCR, CR, VGPR, and PR. There were no significant differences in PFS and OS in the comparison of patients achieving sCR, CR, VGPR or PR. PFS was defined as time from best response achieved until disease progression or death from any cause and was estimated by the Kaplan-Meier method. OS was defined as time from best response achieved until death from any cause.

Figure 2.

Depth of response based on standard and MRD response criteria. PFS (A) and OS (B) of patients stratified according to best response achieved during treatment: MRD, sCR, CR, VGPR, and PR. There were no significant differences in PFS and OS in the comparison of patients achieving sCR, CR, VGPR or PR. PFS was defined as time from best response achieved until disease progression or death from any cause and was estimated by the Kaplan-Meier method. OS was defined as time from best response achieved until death from any cause.

Close modal

The clinical value of serial measurements of the M-protein and sFLC during patient treatment and follow-up is undeniable. Our findings merely exposed the limited utility of standard response criteria to predict different PFS in transplant-eligible MM treated with VRD induction and consolidation. In nontransplant candidates treated with less intensive regimens, such as that of the PETHEMA/GEM CLARIDEX trial, we observed that patients achieving sCR or CR and VGPR had similar outcomes, whereas those in PR displayed inferior PFS (supplemental Figure 4). Consequently, our results urge other groups to investigate these findings in this and other treatment scenarios. If reproducible, such data could support a new iteration of the IMWG 2016 guidelines for response assessment, which may reflect the direct usage of BM aspirates for MRD testing, the standardization of positron emission tomography/computed tomography,23 and the incorporation of novel methods such as mass spectrometry to measure serological response in MM.9,24,25

This study was supported by the Centro de Investigación Biomédica en Red–Área de Oncología-del Instituto de Salud Carlos III (CIBERONC, CB16/12/00369, CB16/12/00400, and CB16/12/00284), Instituto de Salud Carlos III/Subdirección General de Investigación Sanitaria (FIS No. PI15/01956, PI15/02049, FIS PI15/02062, y PI18/01709), Fondo Europeo de Desarrollo Regional (FEDER) and Asociación Española Contra el Cáncer (FCAECC, Predoctoral Grant Junta Provincial Navarra) and Beca Leonardo a Investigadores y Creadores Culturales 2017, Fundación BBVA (IN[17]_BBM_TRA_0236) and internationally by the Cancer Research UK, FCAECC and AIRC under the Accelerator Award Program, Black Swan Research Initiative of the International Myeloma Foundation and the European Research Council (ERC) 2015 Starting Grant (MYELOMANEXT).

Contribution: A.J.-U., B.P., J.S.M., and J.J.L. conceived the analysis; A.J.-U., B.P., M.-V.M., L.R., J. Blade, J.S.M., and J.J.L. designed the analysis protocol; B.P., M.-T.C., and N.P. analyzed the flow cytometry data; A.J.-U., B.P., J.S.M., and J.J.L., analyzed and interpreted the data; A.J.-U. and B.P. performed the statistical analysis; A.J.-U., B.P., J.S.M., and J.J.L. wrote the manuscript; and all authors provided study material or patients and reviewed and approved the manuscript.

Conflict-of-interest disclosure: B.P. reports receiving honoraria for lectures from and membership on advisory boards with Adaptive, Amgen, Bristol-Myers Squibb-Celgene, Creative BioLabs, Janssen, Kite Pharma, Sanofi and Takeda; unrestricted grants from Celgene, EngMab, Roche, Sanofi, and Takeda; and consultancy for Bristol-Myers Squibb-Celgene, Janssen, Sanofi and Takeda. N.P. has received honoraria from Janssen-Cilag, Takeda, and Amgen, and has served in a consulting or advisory role and received travel, accommodations, and expenses from Janssen-Cilag. A.O. has served in a consulting or advisory role for Amgen and Janssen-Cilag. A.S. has received honoraria from Takeda, MSD, BMS/Celgene, Janssen, Amgen, Novartis, Gilead-Kite, Sanofi, Roche, and Alexion; has served in a consulting or advisory role for Takeda, BMS/Celgene, Novartis, Janssen, Gilead Kite, and Sanofi; and has served on the speakers’ bureau for Takeda. L.P. has received honoraria from and served in a consulting or advisory role for Janssen-Cilag and Celgene. M.-V.M. has received honoraria from and has served on the speakers’ bureau for Janssen-Cilag and Celgene. L.R. has received honoraria from Janssen-Cilag and Celgene. J. Blade has received honoraria for lectures and advisory boards from Janssen-Cilag, Celgene, Amgen, and Takeda. J.S.M. has served as a consultant for Bristol-Myers Squibb, Janssen-Cilag, Celgene, Merck, Takeda, Novartis, Amgen, Sanofi, and Roche. J.J.L. has received honoraria for lectures from and has participated in advisory boards for Janssen-Cilag, Celgene, Takeda, Amgen, GSK, and Sanofi. The remaining authors declare no competing financial interests.

A complete list of the members of the GEM/PETHEMA Cooperative Study Group appears in the supplemental appendix.

Correspondence: Juan José Lahuerta, Instituto de Investigación Sanitaria Hospital Universitario 12 de Octubre, Avenida de Córdoba s/n. 28041, Madrid, Spain; e-mail: jjlahuerta@telefonica.net.

Requests for original data should be directed to the corresponding author (jjlahuerta@telefonica.net).

The online version of this article contains a data supplement.

1.
Rosiñol
L
,
Oriol
A
,
Rios
R
, et al
.
Bortezomib, lenalidomide, and dexamethasone as induction therapy prior to autologous transplant in multiple myeloma
.
Blood.
2019
;
138
(
19
):
1337
-
1345
.
2.
Gay
F
,
Cerrato
C
,
Petrucci
MT
, et al
.
Efficacy of carfilzomib lenalidomide dexamethasone (KRd) with or without transplantation in newly diagnosed myeloma according to risk status: Results from the FORTE trial [abstract]
.
J Clin Oncol.
2019
;
138
(
19
). Abstract 8002.
3.
Attal
M
,
Lauwers-Cances
V
,
Hulin
C
, et al;
IFM 2009 Study
.
Lenalidomide, Bortezomib, and Dexamethasone with Transplantation for Myeloma
.
N Engl J Med.
2017
;
138
(
19
):
1311
-
1320
.
4.
Moreau
P
,
Attal
M
,
Hulin
C
, et al
.
Bortezomib, thalidomide, and dexamethasone with or without daratumumab before and after autologous stem-cell transplantation for newly diagnosed multiple myeloma (CASSIOPEIA): a randomised, open-label, phase 3 study
.
Lancet.
2019
;
138
(
19
):
29
-
38
.
5.
Voorhees
PM
,
Kaufman
JL
,
Laubach
J
, et al
.
Daratumumab, lenalidomide, bortezomib, and dexamethasone for transplant-eligible newly diagnosed multiple myeloma: the GRIFFIN trial
.
Blood.
2020
;
138
(
19
):
936
-
945
.
6.
Costa
LJ
,
Chhabra
S
,
Godby
KN
, et al
.
Daratumumab, carfilzomib, lenalidomide and dexamethasone (Dara-KRd) induction, autologous transplantation and post-transplant, response-adapted, measurable residual disease (MRD)-based dara-Krd consolidation in patients with newly diagnosed multiple myelo [abstract]
.
Blood.
2019
;
138
(
19
). Abstract 860.
7.
Kumar
S
,
Paiva
B
,
Anderson
KC
, et al
.
International Myeloma Working Group consensus criteria for response and minimal residual disease assessment in multiple myeloma
.
Lancet Oncol.
2016
;
138
(
19
):
e328
-
e346
.
8.
Paiva
B
,
Puig
N
,
Cedena
M-T
, et al;
GEM (Grupo Español de Mieloma)/PETHEMA (Programa Para el Estudio de la Terapéutica en Hemopatías Malignas) Cooperative Study Group
.
Measurable Residual Disease by Next-Generation Flow Cytometry in Multiple Myeloma
.
J Clin Oncol.
2020
;
138
(
19
):
784
-
792
.
9.
Derman
BA
,
Stefka
AT
,
Jiang
K
, et al
.
Measurable residual disease assessed by mass spectrometry in peripheral blood in multiple myeloma in a phase II trial of carfilzomib, lenalidomide, dexamethasone and autologous stem cell transplantation
.
Blood Cancer J.
2021
;
138
(
19
):
19
.
10.
Mateos
MV
,
Dimopoulos
MA
,
Cavo
M
, et al;
ALCYONE Trial Investigators
.
Daratumumab plus bortezomib, melphalan, and prednisone for untreated myeloma
.
N Engl J Med.
2018
;
138
(
19
):
518
-
528
.
11.
Perrot
A
,
Lauwers-Cances
V
,
Corre
J
, et al
.
Minimal residual disease negativity using deep sequencing is a major prognostic factor in multiple myeloma
.
Blood.
2018
;
138
(
19
):
2456
-
2464
.
12.
Avet-Loiseau
H
,
San-Miguel
J
,
Casneuf
T
, et al
.
Evaluation of Sustained Minimal Residual Disease Negativity With Daratumumab-Combination Regimens in Relapsed and/or Refractory Multiple Myeloma: Analysis of POLLUX and CASTOR
.
J Clin Oncol.
2021
;
138
(
19
):
1139
-
1149
.
13.
Terpos
E
,
Kostopoulos
IV
,
Kastritis
E
, et al
.
Impact of Minimal Residual Disease Detection by Next-Generation Flow Cytometry in Multiple Myeloma Patients with Sustained Complete Remission after Frontline Therapy
.
HemaSphere.
2019
;
138
(
19
):
e300
.
14.
Martinez-Lopez
J
,
Wong
SW
,
Shah
N
, et al
.
Clinical value of measurable residual disease testing for assessing depth, duration, and direction of response in multiple myeloma
[published correction appears in Blood Adv. 2020 Sep 22;4(18):4573.].
Blood Adv.
2020
;
138
(
19
):
3295
-
3301
.
15.
Lahuerta
J-J
,
Jiménez-Ubieto
A
,
Paiva
B
, et al
.
Role of urine immunofixation in the complete response assessment of MM patients other than light-chain-only disease
.
Blood.
2019
;
138
(
19
):
2664
-
2668
.
16.
Bladé
J
,
Samson
D
,
Reece
D
, et al;
Myeloma Subcommittee of the EBMT. European Group for Blood and Marrow Transplant
.
Criteria for evaluating disease response and progression in patients with multiple myeloma treated by high-dose therapy and haemopoietic stem cell transplantation
.
Br J Haematol.
1998
;
138
(
19
):
1115
-
1123
.
17.
Chee
CE
,
Kumar
S
,
Larson
DR
, et al
.
The importance of bone marrow examination in determining complete response to therapy in patients with multiple myeloma
.
Blood.
2009
;
138
(
19
):
2617
-
2618
.
18.
Durie
BG
,
Harousseau
JL
,
Miguel
JS
, et al;
International Myeloma Working Group
.
International uniform response criteria for multiple myeloma [published corrections appear in
Leukemia. 2006;20(12):2220 and 2007;21(5):1134].
Leukemia.
2006
;
138
(
19
):
1467
-
1473
.
19.
Martínez-López
J
,
Paiva
B
,
López-Anglada
L
, et al;
Spanish Multiple Myeloma Group / Program for the Study of Malignant Blood Diseases Therapeutics (GEM / PETHEMA) Cooperative Study Group
.
Critical analysis of the stringent complete response in multiple myeloma: contribution of sFLC and bone marrow clonality
.
Blood.
2015
;
138
(
19
):
858
-
862
.
20.
Lahuerta
J-J
,
Paiva
B
,
Vidriales
M-B
, et al;
GEM (Grupo Español de Mieloma)/PETHEMA (Programa para el Estudio de la Terapéutica en Hemopatías Malignas) Cooperative Study Group
.
Depth of Response in Multiple Myeloma: A Pooled Analysis of Three PETHEMA/GEM Clinical Trials
.
J Clin Oncol.
2017
;
138
(
19
):
2900
-
2910
.
21.
Paiva
B
,
Gutiérrez
NC
,
Rosiñol
L
, et al;
PETHEMA/GEM (Programa para el Estudio de la Terapéutica en Hemopatías Malignas/Grupo Español de Mieloma) Cooperative Study Groups
.
High-risk cytogenetics and persistent minimal residual disease by multiparameter flow cytometry predict unsustained complete response after autologous stem cell transplantation in multiple myeloma
.
Blood.
2012
;
138
(
19
):
687
-
691
.
22.
Paiva
B
,
Vidriales
MB
,
Cerveró
J
, et al;
GEM (Grupo Español de MM)/PETHEMA (Programa para el Estudio de la Terapéutica en Hemopatías Malignas) Cooperative Study Groups
.
Multiparameter flow cytometric remission is the most relevant prognostic factor for multiple myeloma patients who undergo autologous stem cell transplantation
.
Blood.
2008
;
138
(
19
):
4017
-
4023
.
23.
Zamagni
E
,
Nanni
C
,
Dozza
L
, et al
.
Standardization of 18F-FDG-PET/CT According to Deauville Criteria for Metabolic Complete Response Definition in Newly Diagnosed Multiple Myeloma
.
J Clin Oncol.
2021
;
138
(
19
):
116
-
125
.
24.
Mills
JR
,
Barnidge
DR
,
Dispenzieri
A
,
Murray
DL
.
High sensitivity blood-based M-protein detection in sCR patients with multiple myeloma
.
Blood Cancer J.
2017
;
138
(
19
):
e590
.
25.
Puig
N
,
Mateos
M-V
,
Contreras
T
, et al
.
Qip-Mass Spectrometry in High Risk Smoldering Multiple Myeloma Patients Included in the GEM-CESAR Trial: Comparison with Conventional and Minimal Residual Disease IMWG Response Assessment [abstract]
.
Blood.
2019
;
138
(
19
). Abstract
581
.

Author notes

*

A.J-U. and B.P. contributed equally to this study as first authors.

J.S.M. and J.J.L. contributed equally to this study as last authors.

Supplemental data

Sign in via your Institution