• MRD-negativity at 12 months reduced the risk of progression; the treatment effect on MRD was correlated with the treatment effect on PFS.

  • MRD-negativity is reasonably likely to eventually demonstrate a treatment effect on PFS.

Abstract

Estimating progression-free survival (PFS) and overall survival superiority during clinical trials of multiple myeloma (MM) has become increasingly challenging as novel therapeutics have improved patient outcomes. Thus, it is imperative to identify earlier end point surrogates that are predictive of long-term clinical benefit. Minimal residual disease (MRD)-negativity is a common intermediate end point that has shown prognostic value for clinical benefit in MM. This meta-analysis was based on the US Food and Drug Administration guidance for considerations for a meta-analysis of MRD as a clinical end point and evaluates MRD-negativity as an early end point reasonably likely to predict long-term clinical benefit. Eligible studies were phase 2 or 3 randomized controlled clinical trials measuring MRD-negativity as an end point in patients with MM, with follow-up of ≥6 months following an a priori–defined time point of 12 ± 3 months after randomization. Eight newly diagnosed MM studies evaluating 4907 patients were included. Trial-level associations between MRD-negativity and PFS were R2WLSiv, 0.67 (95% confidence interval [CI], 0.43-0.91) and R2copula 0.84 (0.64 to >0.99) at the 12-month time point. The individual-level association between 12-month MRD-negativity and PFS resulted in a global odds ratio (OR) of 4.02 (95% CI, 2.57-5.46). For relapse/refractory MM, there were 4 studies included, and the individual-level association between 12-month MRD-negativity and PFS resulted in a global OR of 7.67 (4.24-11.10). A clinical trial demonstrating a treatment effect on MRD is reasonably likely to eventually demonstrate a treatment effect on PFS, suggesting that MRD may be an early clinical end point reasonably likely to predict clinical benefit in MM, that may be used to support accelerated approval and thereby, expedite the availability of new drugs to patients with MM.

1.
Anderson
KC
,
Auclair
D
,
Adam
SJ
, et al
.
Minimal residual disease in myeloma: application for clinical care and new drug registration
.
Clin Cancer Res
.
2021
;
27
(
19
):
5195
-
5212
.
2.
Ferlay
J
,
Colombet
M
,
Soerjomataram
I
, et al
.
Cancer statistics for the year 2020: an overview
.
Int J Cancer
.
2021
;
149
(
4
):
778
-
789
.
3.
Sung
H
,
Ferlay
J
,
Siegel
RL
, et al
.
Global cancer statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries
.
CA Cancer J Clin
.
2021
;
71
(
3
):
209
-
249
.
4.
Polsinelli
B
,
Tsigkos
S
,
Naumann-Winter
F
,
Mariz
S
,
Sepodes
B
.
Evolving prevalence of haematological malignancies in orphan designation procedures in the European Union
.
Orphanet J Rare Dis
.
2017
;
12
(
1
):
17
.
5.
Turesson
I
,
Bjorkholm
M
,
Blimark
CH
,
Kristinsson
S
,
Velez
R
,
Landgren
O
.
Rapidly changing myeloma epidemiology in the general population: increased incidence, older patients, and longer survival
.
Eur J Haematol
.
2018
;
101
(
2
):
237
-
244
.
6.
SEER
.
Surveillance, epidemiology, and end results program. Cancer stat facts: myeloma
. 2022. Accessed 5 June 2024. https://seer.cancer.gov/statfacts/html/mulmy.html.
7.
Holstein
SA
,
Suman
VJ
,
McCarthy
PL
.
Should overall survival remain an endpoint for multiple myeloma trials?
.
Curr Hematol Malig Rep
.
2019
;
14
(
1
):
31
-
38
.
8.
Abrams Kaplan
D
.
Multiple myeloma: top 10 advances in the past 10 years
.
Target Ther Oncol
.
2022
;
11
(
4
):
70
.
9.
Avet-Loiseau
H
,
Ludwig
H
,
Landgren
O
, et al
.
Minimal residual disease status as a surrogate endpoint for progression-free survival in newly diagnosed multiple myeloma studies: a meta-analysis
.
Clin Lymphoma Myeloma Leuk
.
2020
;
20
(
1
):
e30
-
e37
.
10.
Bahlis
NJ
,
Dimopoulos
MA
,
White
DJ
, et al
.
Daratumumab plus lenalidomide and dexamethasone in relapsed/refractory multiple myeloma: extended follow-up of POLLUX, a randomized, open-label, phase 3 study
.
Leukemia
.
2020
;
34
(
7
):
1875
-
1884
.
11.
Costa
LJ
,
Chhabra
S
,
Medvedova
E
, et al
.
Daratumumab, carfilzomib, lenalidomide, and dexamethasone with minimal residual disease response-adapted therapy in newly diagnosed multiple myeloma
.
J Clin Oncol
.
2022
;
40
(
25
):
2901
-
2912
.
12.
Landgren
O
,
Iskander
K
.
Modern multiple myeloma therapy: deep, sustained treatment response and good clinical outcomes
.
J Intern Med
.
2017
;
281
(
4
):
365
-
382
.
13.
San-Miguel
J
,
Avet-Loiseau
H
,
Paiva
B
, et al
.
Sustained minimal residual disease negativity in newly diagnosed multiple myeloma and the impact of daratumumab in MAIA and ALCYONE
.
Blood
.
2022
;
139
(
4
):
492
-
501
.
14.
Landgren
O
,
Gormley
N
,
Turley
D
, et al
.
Flow cytometry detection of minimal residual disease in multiple myeloma: lessons learned at FDA-NCI roundtable symposium
.
Am J Hematol
.
2014
;
89
(
12
):
1159
-
1160
.
15.
Munshi
NC
,
Avet-Loiseau
H
,
Anderson
KC
, et al
.
A large meta-analysis establishes the role of MRD negativity in long-term survival outcomes in patients with multiple myeloma
.
Blood Adv
.
2020
;
4
(
23
):
5988
-
5999
.
16.
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
;
17
(
8
):
e328
-
e346
.
17.
Kumar
SK
,
Callander
NS
,
Adekola
K
, et al
.
NCCN clinical practice guidelines in oncology: multiple myeloma, version 3.2021
.
J Natl Compr Canc Netw
.
2020
;
18
(
12
):
1685
-
1717
.
18.
HHS, FDA, OCE, CDER, CBER
.
Hematologic Malignancies: Regulatory Considerations for Use of Minimal Residual Disease in Development of Drug and Biological Products for Treatment. Guidance for Industry. FDA-2018-D-3090
. January 2020. Accessed 5 June 2024. https://www.fda.gov/regulatory-information/search-fda-guidance-documents/hematologic-malignancies-regulatory-considerations-use-minimal-residual-disease-development-drug-and.
19.
Cavo
M
,
San-Miguel
J
,
Usmani
SZ
, et al
.
Prognostic value of minimal residual disease negativity in myeloma: combined analysis of POLLUX, CASTOR, ALCYONE, and MAIA
.
Blood
.
2022
;
139
(
6
):
835
-
844
.
20.
Landgren
O
,
Devlin
S
,
Boulad
M
,
Mailankody
S
.
Role of MRD status in relation to clinical outcomes in newly diagnosed multiple myeloma patients: a meta-analysis
.
Bone Marrow Transplant
.
2016
;
51
(
12
):
1565
-
1568
.
21.
Munshi
NC
,
Avet-Loiseau
H
,
Rawstron
AC
, et al
.
Association of minimal residual disease with superior survival outcomes in patients with multiple myeloma: a meta-analysis
.
JAMA Oncol
.
2017
;
3
(
1
):
28
-
35
.
22.
Paiva
B
,
Zherniakova
A
,
Nuñez-Córdoba
JM
, et al
.
Impact of treatment effect on MRD and PFS: an aggregate data analysis from randomized clinical trials in multiple myeloma
.
Blood Adv
.
2024
;
8
(
1
):
219
-
223
.
23.
Voorhees
PM
,
Sborov
DW
,
Laubach
J
, et al
.
Addition of daratumumab to lenalidomide, bortezomib, and dexamethasone for transplantation-eligible patients with newly diagnosed multiple myeloma (GRIFFIN): final analysis of an open-label, randomised, phase 2 trial
.
Lancet Haematol
.
2023
;
10
(
10
):
e825
-
e837
.
24.
Roussel
M
,
Moreau
P
,
Hebraud
B
, et al
.
Bortezomib, thalidomide, and dexamethasone with or without daratumumab for transplantation-eligible patients with newly diagnosed multiple myeloma (CASSIOPEIA): health-related quality of life outcomes of a randomised, open-label, phase 3 trial
.
Lancet Haematol
.
2020
;
7
(
12
):
e874
-
e883
.
25.
Moreau
P
,
Hulin
C
,
Perrot
A
, et al
.
Maintenance with daratumumab or observation following treatment with bortezomib, thalidomide, and dexamethasone with or without daratumumab and autologous stem-cell transplant in patients with newly diagnosed multiple myeloma (CASSIOPEIA): an open-label, randomised, phase 3 trial
.
Lancet Oncol
.
2021
;
22
(
10
):
1378
-
1390
.
26.
Mateos
M-V
,
Dimopoulos
MA
,
Cavo
M
, et al
.
Daratumumab plus bortezomib, melphalan, and prednisone for untreated myeloma
.
N Engl J Med
.
2018
;
378
(
6
):
518
-
528
.
27.
Mateos
M-V
,
Cavo
M
,
Blade
J
, et al
.
Overall survival with daratumumab, bortezomib, melphalan, and prednisone in newly diagnosed multiple myeloma (ALCYONE): a randomised, open-label, phase 3 trial
.
Lancet
.
2020
;
395
(
10218
):
132
-
141
.
28.
Burzykowski
T
,
Molenberghs
G
,
Buyse
M
.
The validation of surrogate end points by using data from randomized clinical trials: a case-study in advanced colorectal cancer
.
J R Stat Soc
.
2004
;
167
(
1
):
103
-
124
.
29.
Paiva
B
,
Gutierrez
NC
,
Rosinol
L
, et al
.
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
;
119
(
3
):
687
-
691
.
30.
Paiva
B
,
Martinez-Lopez
J
,
Vidriales
MB
, et al
.
Comparison of immunofixation, serum free light chain, and immunophenotyping for response evaluation and prognostication in multiple myeloma
.
J Clin Oncol
.
2011
;
29
(
12
):
1627
-
1633
.
31.
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
;
112
(
10
):
4017
-
4023
.
32.
Hultcrantz
M
,
Rustad
EH
,
Yellapantula
V
, et al
.
Capture rate of V(D)J sequencing for minimal residual disease detection in multiple myeloma
.
Clin Cancer Res
.
2022
;
28
(
10
):
2160
-
2166
.
You do not currently have access to this content.
Sign in via your Institution