Oncology

Multiple Myeloma

Advertisment

Minimal Residual Disease Testing in Multiple Myeloma

expert roundtables by Kenneth C. Anderson, MD; Sagar Lonial, MD, FACP; S. Vincent Rajkumar, MD
Overview

Minimal residual disease (MRD) negativity can be achieved and has clear prognostic value in multiple myeloma. However, efforts to understand how best to use MRD testing clinically are still ongoing.

QUESTION:
How has MRD assessment evolved in recent years, and what unique insights does such testing provide?
“The achievement of MRD negativity is no longer a dream, which is exciting, but that is not enough. Rather, the durability of MRD negativity is also important.”
— Kenneth C. Anderson, MD

Getting fewer than 1 in 100,000 or 1 in 1,000,000 multiple myeloma cells in the bone marrow after treatment is very exciting. In the past, we have only been able to achieve that with allogeneic transplantation, but now we can achieve MRD negativity at a high frequency with the advent of novel treatments. A large meta-analysis showed that MRD negativity confers prolonged progression-free survival (PFS) and overall survival (OS) advantages in patients with newly diagnosed multiple myeloma or in those with relapsed/refractory multiple myeloma.

 

The biologic excitement is that if you can get to MRD negativity and get rid of the abnormal multiple myeloma clone early, you can potentially prevent the evolution that inevitably occurs in multiple myeloma and leads to disease resistance and, ultimately, relapse. The achievement of MRD negativity is no longer a dream, which is exciting, but that is not enough. Rather, the durability of MRD negativity is also important. Persistent MRD negativity is associated with a long-term benefit after treatment in newly diagnosed, transplant-eligible and -ineligible patients, as well as after chimeric antigen receptor T-cell therapy for multiply relapsed disease.

 

Another potential application of MRD analysis that is being studied is in the context of using MRD to guide post-transplant consolidation and possible treatment cessation. The current paradigm is lifelong maintenance therapy. Data from the phase 2 MASTER trial showed that the 2-year cumulative risk for progression for patients with 0, 1, or 2 or more high-risk cytogenetic abnormalities was 9%, 9%, and 47%, respectively. Such results demonstrate that, while we are excited that we can achieve MRD negativity, we are still learning how to use MRD negativity clinically.

 

Another hope with MRD negativity is that it can be used as a regulatory end point in appropriately designed clinical trials. Unprecedented PFS is now being observed in newly diagnosed and relapsed/refractory multiple myeloma, and, unless alternative end points to PFS are identified, new studies will not have a readout for many years, which may be too late for many of our patients.

 

One final thought is to emphasize the importance of assessing MRD negativity inside and outside the bone marrow. We need to be sure that we also have imaging negativity (eg, positron emission tomography/computed tomography or magnetic resonance imaging) when we assess MRD negativity, as defined in the International Myeloma Working Group criteria.

"I think that the focus is appropriately on MRD in high-risk patients in the sense that we are really keen on getting high-risk patients to an MRD-negative status. The higher the patient’s risk, the more we want to find a way to modify therapy to get them to MRD negativity.”
— S. Vincent Rajkumar, MD

MRD cutoffs for prognosis have become clearer, and it seems that, if prognostic information is the goal, then 10-6 seems much better for predicting outcomes than 10-5. It is also important to emphasize that a single result of MRD negativity has very little meaning compared with a sustained MRD negativity report across multiple time points. If we are going to use MRD to guide treatment decisions such as treatment discontinuation, we need to have multiple MRD-negative readouts because that has more prognostic power than a single reading.

 

We are beginning to explore new ways to estimate MRD. Next-generation sequencing and next-generation flow cytometry are methods that are used with bone marrow samples, but there is also increasing interest in alternative methods that might allow us to reduce the need for more frequent bone marrow biopsies. In that regard, very sensitive mass spectrometry or other genomic methods might be used for MRD testing. Interesting results have been reported with mass spectrometry–based methodologies to predict survival outcomes, but further investigation is needed.

 

It is difficult to use early MRD assessments at 4 and 6 months to determine long-term outcomes. I think that the focus is appropriately on MRD in high-risk patients in the sense that we are really keen on getting high-risk patients to an MRD-negative status. The higher the patient’s risk, the more we want to find a way to modify therapy to get them to MRD negativity. While this is not fully data driven, it is based on our philosophy that these patients do not do well unless they become MRD negative.

“In many cases, MRD negativity may give us more information about the biology and responsiveness of the disease to a given therapy than the curative potential of the therapy. . . . Thus, I see MRD as another variable that we need to understand.”
— Sagar Lonial, MD, FACP

There are many studies in which patients who achieve MRD negativity have a longer PFS and OS than patients who do not. And it is absolutely true that achieving such a low disease burden correlates with longer PFS, much like achieving a complete response correlated with better survival 10 years ago. However, we also know that many patients who achieve MRD negativity will ultimately relapse. In many cases, MRD negativity may give us more information about the biology and responsiveness of the disease to a given therapy than the curative potential of the therapy.

 

Thus, I see MRD as another variable that we need to understand. To even consider discontinuation, I think that we would need to use 10-6 as the cutoff and to see MRD negativity sustained for 12 months or longer. For patients with multiple myeloma who are at standard risk and receive lenalidomide, bortezomib, and dexamethasone in combination with autologous stem cell transplantation followed by lenalidomide maintenance, the estimated median PFS is 76.5 months. So, short-term end points such as MRD testing at cycle 4 or 8 pale in comparison to the follow-ups after 50 months that we see with the current continuous therapy model.

References

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. doi:10.1158/1078-0432.CCR-21-1059

 

ClinicalTrials.gov. Non-invasive MRD assessment in multiple myeloma (NIRVANA). Updated November 23, 2022. Accessed October 6, 2023. https://clinicaltrials.gov/study/NCT05625971

 

Cohen AD, Mateos M-V, Cohen YC, et al. Efficacy and safety of cilta-cel in patients with progressive multiple myeloma after exposure to other BCMA-targeting agents. Blood. 2023;141(3):219-230. doi:10.1182/blood.2022015526

 

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. doi:10.1200/JCO.21.01935

 

Costa LJ, Chhabra S, Medvedova E, et al. Minimal residual disease response-adapted therapy in newly diagnosed multiple myeloma (MASTER): final report of the multicentre, single-arm, phase 2 trial. Lancet Haematol. 2023 Sep 27:S2352-3026(23)00236-3. doi:10.1016/S2352-3026(23)00236-3

 

Costa L, Medvedova E, Chhabra S, et al. Quadruplet induction therapy, ASCT and MRD-modulated consolidation and treatment cessation in newly diagnosed multiple myeloma: final analysis of the MASTER trial [abstract S203]. Abstract presented at: 2023 European Hematology Association Congress; June 11, 2023; Frankfurt, Germany.

 

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;11(2):19. doi:10.1038/s41408-021-00418-2

 

Dispenzieri A, Krishnan A, Arendt B, et al. Mass-Fix better predicts for PFS and OS than standard methods among multiple myeloma patients participating on the STAMINA trial (BMT CTN 0702 /07LT). Blood Cancer J. 2022;12(2):27. doi:10.1038/s41408-022-00624-6

 

Joseph NS, Kaufman JL, Dhodapkar MV, et al. Long-term follow-up results of lenalidomide, bortezomib, and dexamethasone induction therapy and risk-adapted maintenance approach in newly diagnosed multiple myeloma. J Clin Oncol. 2020;38(17):1928-1937. doi:10.1200/JCO.19.02515

 

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. doi:10.1016/S1470-2045(16)30206-6

 

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. doi:10.1182/bloodadvances.2020002827

 

Munshi NC, Paiva B, Martin T, et al. Efficacy outcomes and characteristics of patients with multiple myeloma (MM) who achieved sustained minimal residual disease negativity after treatment with ciltacabtagene autoleucel (cilta-cel) in CARTITUDE-1 [abstract 2030]. Abstract presented at: 64th American Society of Hematology Annual Meeting and Exposition; December 10-13, 2022; New Orleans, LA.

 

Paiva B, Manrique I, Dimopoulos MA, et al. MRD dynamics during maintenance for improved prognostication of 1280 patients with myeloma patients in the TOURMALINE-MM3 and -MM4 trials. Blood. 2023;141(6):579-591. doi:10.1182/blood.2022016782.

 

Paiva B, Manrique I, Rytlewski JA, et al. Early and sustained undetectable measurable residual disease (MRD) after idecabtagene vicleucel (ide-cel) defines a subset of multiple myeloma (MM) patients in KarMMa achieving prolonged survival. Blood. 2022;140(suppl 1):2104-2105. doi:10.1182/blood-2022-166658

 

Paiva B, Zherniakova A, Nuñez-Córdoba JM, et al. Impact of treatment effect on MRD and PFS: an aggregate analysis from randomized clinical trials in multiple myeloma. Blood Adv. 2023 Aug 28:bloodadvances.2023010821. doi:10.1182/bloodadvances.2023010821

Kenneth C. Anderson, MD

Kraft Family Professor of Medicine
Harvard Medical School
Director, LeBow Institute for Myeloma Therapeutics
Director, Jerome Lipper Multiple Myeloma Center
Dana-Farber Cancer Institute
Boston, MA

Sagar Lonial, MD, FACP

Professor and Chair
Department of Hematology and Medical Oncology
Chief Medical Officer
Winship Cancer Institute
Emory University School of Medicine
Atlanta, GA

S. Vincent Rajkumar, MD

Edward W. and Betty Knight Scripps Professor of Medicine
Mayo Clinic
Rochester, MN

Advertisment