Journal List > Lab Med Online > v.14(3) > 1516087637

Measurable Residual Disease of Multiple Myeloma: Current Methods and Future Directions

Abstract

The detection of measurable residual disease (MRD) has a significant impact on the prognosis of patients with multiple myeloma (MM). While complete response rates have increased due to the discovery of novel agents and hematopoietic stem cell transplantation, a significant number of patients experience relapse after achieving a complete response. Therefore, it is necessary to evaluate the deeper response to identify patients who are at a higher risk of early relapse. Patients who achieve MRD negativity through highly sensitive MRD assays have shown improved survival. Moreover, MRD assays with sensitivities greater than 10-5 have been included in various clinical trials. This review focuses on high-sensitivity flow cytometry and next-generation sequencing (NGS)-based MRD in MM. In this article, the current MRD approaches in MM have been summarized, considering the specific characteristics of each assay and harmonization to overcome substantial heterogeneity. Furthermore, we discuss the issues in MM MRD that require further investigation.

초록

다발골수종에서 측정가능잔존질환의 검출은 환자의 예후에 상당한 영향을 미친다. 다발골수종의 치료 과정에 신약과 조혈모세포이식이 도입되면서 반응 평가에서 완전 관해 비율이 과거에 비해 증가하였지만, 완전 관해에 이른 환자 중 상당수가 재발을 경험한다. 재발 가능성이 높은 고위험군 환자를 조기에 발견하기 위해서는 기존보다 더 깊은 반응 단계까지 평가해야 한다. 여러 연구에서, 전통적인 검사법에 비해 훨씬 더 민감한 검사법을 사용하여 측정가능잔존질환을 검출하였을 때 음성으로 평가받은 환자들이 분명한 예후 개선 효과를 보였다고 보고하였다. 따라서 10-5 이하의 잔존질환까지 측정할 수 있는 민감한 검사법이 여러 임상시험에도 포함되었다. 본 종설에서는 다발골수종에서 유세포분석법과 차세대염기서열분석 기반의 매우 민감한 측정가능잔존질환 검사에 초점을 맞추어 기술하고자 한다. 다발골수종에서 현재 적용되고 있는 잔존질환 측정 검사법의 종류, 각 검사법의 구체적인 특성 및 검사법 간 차이에서 비롯되는 이질성을 극복하기 위하여 시도되고 있는 측정방법 일치화 전략에 대해 기술하였다. 또 다발골수종의 잔존질환 검출 분야에서 향후 추가적인 연구를 요하는 주제에 대해서도 언급하였다.

INTRODUCTION

In recent years, there has been increasing emphasis on the prognostic value of measurable residual disease (MRD) detection in multiple myeloma (MM). The standard response criteria proposed by the International Myeloma Working Group (IMWG) primarily rely on reductions in serum monoclonal (M)-protein, serum free light chain ratio, and quantification of clonal plasma cells in bone marrow (BM) biopsy through immunohistochemistry [1]. Therefore, it is recommended to use additional methods for detecting residual disease, especially in patients who achieve a very good partial response (VGPR) or complete response (CR). This recommendation stems from previous studies that have shown an association between MRD negativity and improved overall survival (OS) outcomes [2, 3]. Currently, MM MRD assessments are widely conducted in numerous clinical trials, with MRD negativity serving as a surrogate biomarker for progression-free survival (PFS) and OS acting as an endpoint marker for assessing treatment response [4]. A meta-analysis demonstrated that the achievement of MRD negativity is associated with improved OS outcomes in patients with transplant-eligible newly diagnosed MM and transplant-ineligible and relapsed/refractory MM, regardless of the MRD assessment methods used [5]. The analysis revealed a relationship between survival outcomes and increasing MRD sensitivity thresholds, with MRD negativity at thresholds of 10-6, 10-5, and 10-4 showing survival differences.
Traditionally, 4 to 8 color fiow cytometry was used to evaluate MRD in MM; however, sometimes allele-specific oligonucleotide quantitative PCR (ASOqPCR) was used to evaluate MRD in MM [6-8]. These studies have confirmed the relevance of MRD measurement for the identification of patients with MM at a higher risk of relapse. Even though CR rates have increased because of the introduction of multidrug combinations and autologous hematopoietic stem cell transplantation (ASCT), a significant number of patients still experience relapse [1]. Therefore, the need for more sensitive MRD approaches has increased. In 2017, the EuroFlow consortium published a standardized high-sensitivity multicolor fiow cytometry approach known as next-generation fiow (NGF) MRD, which improves sensitivity up to 10-5 and is applicable for MRD monitoring in MM [9]. However, regarding molecular testing, ASOqPCR has limitations, as it requires specific primer sets for each patient and separates PCR amplification products by length rather than by the actual DNA sequences. Moreover, the use of NGS in hematologic malignancies has increased, leading to the transition of molecular MRD assessment to NGS-based clonality assays, and NGS for immunoglobulin rearrangements has been applied to MM MRD [10, 11]. The type of MRD method to be used in a real-world setting depends on factors such as feasibility, clinical demand, the expertise required for performing and interpreting the analysis, and the availability of instruments. Moreover, achieving higher sensitivity is more important than selecting a specific method. While a deep response with a sensitivity of 10-6 is ideal, a sensitivity of at least 10-5 is needed for MM MRD [1, 12].
In this review, the aim is to provide a summary of the current MRD methodologies incorporated in MM and discuss the future directions of MRD assessment in MM.

MRD ASSESSMENT METHODS

1. High-sensitivity flow cytometric MRD assessment

The IMWG consensus response criteria for “fiow MRD-negative” requires a complete response and the absence of aberrant clonal plasma cells on BM aspirates using the EuroFlow-based NGF or an equivalent validated method with a minimum sensitivity of 1 in 105 nucleated cells or higher [1]. This high-sensitivity NGF method includes an optimized two-tube 8-color antibody combination and adopts the bulk-lysis procedure as a reference for cell acquisition of ≥107. Moreover, it consists of six backbone fluorochrome-conjugated antibodies: CD38-FITC, CD56-PE, CD45-PerCPCy5.5, CD19-PECy7, CD138-BV421, and CD27-BV510. Each tube also contains additional fiuorochrome-conjugated antibodies: tube 1 includes CD117-APC and CD81-APC-C750, while tube 2 includes cytIgKappa-APC and cytIgLambda-APC-C750 [9]. This method incorporates automated gating and identification of plasma cells against a reference database of normal and patient BM. Since the IMWG response criteria require EuroFlow-based NGF or an equivalent validated method, several studies have reported alternative high-sensitivity fiow MRD methods [13-16]. For example, Roshal et al. performed a comparative study between a single 10-color tube and an 8-color 2-tube panel validated by the EuroFlow group [14]. A single-tube, 9-color panel assay was validated with CD200 added and surface staining only. Through an established validation process, this streamlined approach showed a detection level of 10-5 and wide applicability [15]. Efforts to find a more efficient way for NGF MRD analysis include panel composition and the sample preparation method since it is one of the adjustable pre-analysis factors. Bayly et al. compared the post-lysis preparation technique with a modified pre-lysis technique from the EuroFlow method by reducing the number of washing steps [16]. A two-tube, 8-color panel was chosen with CD200 and CD3 added. The modified alternative approach showed equivalent performance, with no significant difference in the cellular composition between the samples. These validated approaches expand available options and contribute to the wider application of NGF MRD analysis.
Since fiow MRD consists of variations in panel selection, sample preparation, and data analysis, there is significant heterogeneity in the methodology, assessment, and reporting of the result. To adopt fiow MRD results as a surrogate endpoint across countries, laboratories, and various clinical trials, international harmonization efforts were proposed for data analysis and MM MRD using fiow cytometry [12, 17]. Soh et al. reported an international harmonized approach for a consensus algorithm by high-sensitivity fiow cytometry, including specific gating strategies, cell enumeration, and evaluation of sample adequacy [17]. A consensus gating protocol was designed, especially for the enumeration of “total analyzed cells,” which affects the calculation of the limit of detection (LOD) and lower limit of quantitation (LLOQ). In the harmonized gating approach, the first step was to exclude debris and doublets and identify singlets. The total number of analyzed cells included CD45+ leukocytes, CD45 erythrocytes, and CD45−/dim aberrant plasma cells. Next, total plasma cells expressing bright CD38+, CD138+, and CD45+/− were identified using bivariate plots of CD38, CD138, and CD45. After which, considering the typical expression pattern of normal plasma cells, bright for CD38, positive for CD19, CD27, CD45, CD81, and negative for CD56, CD117, and polytypic cytoplasmic light chains, aberrant expression of abnormal plasma cells was considered. The presence of mast cells, erythroid precursors, myeloid precursors, or hematogones has been suggested as parameters of hemodilution in MRD samples [17]. When comparing the measurement of MM MRD between independent analysis using the in-house analysis method and the outcome after the adoption of the harmonized approach, the overall consistency between participants improved after adopting the consensus approach, especially in the enumeration of “total analyzed cells,” calculation of LOD and LLOQ, and in defining sample adequacy. Moreover, implementing a suitable harmonized approach diminishes interlaboratory variation, thereby enabling the application of fiow MRD assessment for the evaluation of MM disease status in a real-world setting.
As targeted immunotherapy using anti-CD38 monoclonal antibodies emerged, high-sensitivity fiow MRD approaches faced difficulties in plasma cell identification. While positive expression of CD38 and CD138 and decreased expression of CD45 are important in discriminating plasma cells from other lymphocytes, certain antibodies cannot be used as a gating marker occasionally. CD138 expression-downregulated subpopulations of abnormal plasma cells exist, especially in MM cells from the hypoxic bone marrow microenvironment [18]. Decreased CD38 positivity in the plasma cells was observed after immunotherapy and treatment with daratumumab [19]. Therefore, alternative plasma cell gating strategies have been proposed, including the CD38 multiepitope antibody incorporated in EuroFlow, the CD38 nanobody, the intracytoplasmic marker VS38c, CD319, and CD229 [19-22]. Moreover, VS38c recognizes a transmembrane protein on the rough endoplasmic reticulum and helps distinguish plasma cells from other hematopoietic cells, including polytypic B cells. Clonal plasma cells from patients with recent daratumumab exposure exhibit dim CD38 expression, while VS38c expression remains high. Antibody combinations, including alternative plasma cell markers CD319 and CD229, showed considerable resolution in discerning abnormal plasma cells, even in the plasma cells treated with an anti-CD319 monoclonal antibody. Although these alternative markers require each laboratory to redesign the current panel, clinical laboratories performing MM MRD analysis should consider the impact of these targeted immunotherapies when interpreting the results of treated specimens.

2. NGS-based MRD assessment

According to the IMWG response criteria, “sequencing MRD-negative” is defined as the absence of clonal plasma cells on BM aspirates using NGS, specifically via the LymphoSIGHT platform (currently ClonoSEQ, Adaptive Biotechnologies, WA) or an equivalent validated method with a minimum sensitivity of 1 in 105 nucleated cells or higher, in addition to achieving a complete response [1, 23]. NGS-based MRD assessment focuses on the third complementarity-determining region (CDR3) at the junction of variable (V), diversity (D), and joining (J) immunoglobulin gene regions, particularly in the immunoglobulin heavy locus (IGH) gene [24]. To avoid the omission of clonotypes due to amplification bias and indistinct results against the background of polyclonal B cells, primer sets include the IGH frameworks 1, 2, and 3 (FR1, FR2, FR3), along with the upstream leader sequence region. Moreover, IGK (immunoglobulin kappa locus) and IGL (immunoglobulin lambda locus) can be used to track clonotypes. However, the two light chain genes are considered less reliable markers than IGH as light chain rearrangements involve only V and J regions and lack D segments [24].
Diverse genetic events result in patient-specific patterns through VDJ rearrangement, enabling the tracking of abnormal plasma cells. The sequence variations of clonal rearrangement provide genetic information for individual molecular profiling and are rarely affected by immunophenotypic changes due to immunotherapy [25-27]. Furthermore, analyzing serial samples allows for the investigation of clonal evolution. Moreover, NGS-based MRD can be applied to samples under various conditions, such as stored blood samples and formalin-fixed paraffin-embedded (FFPE) tissue. However, using good-quality FFPE samples with an appropriate fixation method, processing protocols, and a relatively short storage period is crucial to obtaining interpretable results for clinical diagnosis and monitoring [28]. In addition, NGS-based MRD assays can achieve a sensitivity of 10-5 using a relatively low number of cell inputs. With the wide availability of NGS, many clinical laboratories are introducing NGS MRD testing. However, NGS-based MRD requires mandatory initial sample testing to identify patient-specific clones. A risk of trackable clone detection failure exists mainly due to sample quality, such as a low clonal plasma cell burden and hemodilution, and ineffective primer annealing problems caused by somatic hypermutation. These detection failure rates of trackable clones have been reported to be as high as 20%, especially when using the FR1 assay alone in patients with MM [28, 29]. However, the use of several methods can improve success rates. Firstly, incorporating more primer sets such as leader, FR2, and FR3 and combining them with the IGK assay enables clonal detection rates higher than 95% in all B-cell neoplasms, including MM. The addition of leader primers significantly increases the clonal detection rate, similar to the detection rate of assays using a combination of FR1, FR2, and FR3 [28]. Secondly, using an adequate-quality BM sample with a high tumor cell burden decreases the failure rate of identifying clonal sequences. Enrichment of CD138+ tumor cells is recommended.
ClonoSEQ has been approved by the United States Food and Drug Administration; however, it is not available in Korea as it requires samples to be sent to the central laboratory outside of Korea. On the contrary, the LymphoTrack assay (Invivoscribe, San Diego, CA) is a commercially available NGS-based clonality assay available in Korea for in-house sequencing and has been tested in MM [30]. The EuroClonality-NGS Consortium reported a standardized NGS panel assay to detect immunoglobulin and T-cell receptor gene rearrangements for clonality assessment [31, 32], and it is expected to be used for MRD analysis of various lymphoid neoplasms, including MM.
The prognostic values of MRD negativity using the ClonoSEQ assay have been proven in several clinical trials [33-35]. Patients who achieved CR and MRD negativity showed significantly better PFS than those who failed to reach CR or were MRD positive. Ching et al. demonstrated the ability of the clonoSEQ to detect MRD at a level of 10-6 by serial dilution experiments, which showed a potential for a highly sensitive tool for the diagnosis and monitoring of lymphoid malignancies, including MM [23, 36]. Baseline identification of trackable clones and a validation study encompassing the establishment of LOD, linearity, precision, and reproducibility for the lymphoTRACK assay have been performed in previous studies [28, 37]. Medina et al. showed that MM plasma cells have different characteristics from other mature B-cell malignancies such as chronic lymphocytic leukemia and mantle cell lymphoma in the distribution of IGH gene repertoires [38]. In previous studies on Koreans with MM, the clonality detection rate was between 88–95% using the IGH FR1 assay and/or IGK assay and primer sets available in Korea [25, 30]. The profile of IGH and IGK gene usage in Koreans with MM was similar to those observed in previous studies conducted in the Western population. However, there were certain differences in the distribution of specific IGHV genes and the IGK rearrangement. These differences could be attributed to ethnicity-based variations, which need to be further investigated through future studies involving a larger number of Korean patients with MM [25].

3. Comparison of techniques

As previously mentioned, the IMWG response criteria do not recommend a specific MRD method but emphasize the importance of using a validated assay with sufficiently high sensitivity [1]. Previous comparison studies between NGF and NGS-based MRD assays demonstrate a good correlation and a high concordance rate ranging from 80% to 93% [29, 39-42]. While there have been certain discordant cases, including MRD-positive cases detected only by NGF or NGS, most of these cases had MRD levels below 10-5, suggesting a difference in sensitivity between the two methods or differences in the sampling procedure [28, 29, 39]. Sample quality may provide a few possible explanations, as it significantly impacts the results of both MRD methods, also related to the burden of plasma cells. Typically, the best-quality sample from the first pull of BM aspirate is used for cytomorphology, while subsequent samples with a risk of hemodilution are used for MRD analysis [29]. Differences in results may occur when comparing the highest-quality sample with other specimens that may have experienced hemodilution.
Moreover, the MRD status obtained from both methods is significantly associated with the PFS and OS of patients with MM [5, 39, 43]. In addition, Medina et al. evaluated the clinical significance of cases where both NGF and NGS assays showed negativity for MRD and discordant cases [39]. They observed no statistical differences in three-year PFS between the double-negative and discordant cases, although both subgroups had significantly better PFS rates compared to the double-negative cases. Ho et al. reported the risk of relapse in double-negative and discordant cases [29]. Approximately 20% of patients with double negativity using both NGF and NGS assays showed evidence of relapse within a median follow-up time of 36.3 months. Among the twelve discordant cases, three patients showed a relapse, and MRD was identified at a low level, near the limit of detection. When interpreting these low-level MRD results and evaluating their prognostic impact, sampling differences should be taken into consideration.
The characteristics of the NGF and NGS methods are compared in Table 1. NGF-based MRD analysis offers robust advantages, including increased sensitivity compared to conventional flow MRD, a fast turnaround time, does not require baseline sample testing, and can evaluate sample quality and the normal B-cell compartment by identifying hematopoietic cells other than plasma cells, such as mast cells, B-cell precursors, and erythroblasts. However, there are certain disadvantages. Additionally, it requires fresh samples, i.e., within 48 hours of obtaining the sample, to acquire viable cells, cannot be performed on FFPE tissue, and necessitates experienced personnel to carry out the procedure [9, 13, 44, 45]. On the contrary, NGS-based MRD assays can detect low levels of tumor cells and can be applied to various sample types other than blood. Moreover, it is minimally affected by immunotherapy, such as anti-CD38 monoclonal antibody treatment. Serial monitoring through NGS MRD enables the acquisition of valuable information regarding patient-specific clonotypes and clonal evolution. However, it requires baseline sample testing to identify patient-specific rearrangements, has a risk of detection failure for trackable clones, and needs a relatively longer turnaround time and is expensive [26, 39, 44]. However, the cost of MRD assessment using NGF or NGS assays varies between countries, hospitals, and laboratories and is greatly infiuenced by reimbursement policies [13, 26, 44]. Therefore, clinical laboratories should choose suitable MRD methods based on their availability and potential needs.

4. Challenges and future directions

The field of MRD methods, procedures, assay sensitivity, evaluation, and reporting processes is highly heterogeneous. Standardization of the analytical aspects of NGF and NGS is necessary. However, achieving complete standardization can be challenging due to the need to unify methodologies. To promote international harmonization of MRD evaluation in clinical trials for MM, Costa et al. proposed several consensus statements [12]. The panelists provided recommendations on various topics, including analytical requirements, assay performance, BM sampling, and the timing of MRD testing in clinical trials. These consensus statements pertain to technical aspects and include the following criteria: the analytical performance of the assay must be validated with a defined limit of blank (LOB), LOQ, and LOD. MRD assays should achieve a LOD <10-5 and should apply to over 90% of patients. If possible, sensitivity levels below 10-6 should be reported. To ensure sufficient sensitivity, it is preferable to perform the MRD assay using the first pull of the BM aspirate.
The feasibility of MRD in peripheral blood (PB) was suggested [12]. MM is characterized by multiple lytic lesions and extramedullary involvement, suggesting the spread of abnormal plasma cells from the primary tumor to various BM niches through peripheral circulation [46]. High-sensitivity NGF can detect circulating tumor plasma cells (CTPCs) [47], allowing for monitoring of disease status using PB instead of BM specimens. This approach reduces the burden on patients, sparing them from painful procedures and facilitating periodic testing when needed. Furthermore, PB can be used in cases of poor BM aspirate quality due to patchy infiltration. Previous reports have highlighted the prognostic impact of CTPCs, although they have indicated that MRD detection in PB is less sensitive than in BM [48-50]. Nonetheless, MRD assessment with PB has the potential to complement BM-based MRD analysis. However, further investigation and cross-validation with BM-based MRD assays are required.

CONCLUSION

MM MRD is currently incorporated into various clinical trials and infiuences clinical decision-making. A conventional complete response is no longer considered sufficient to assess disease status and prognostication. Detecting deep-level residual disease is increasingly crucial for identifying higher-risk patients and predicting early relapse. Well-validated, highly sensitive MRD assays, including high-sensitivity NGF, NGS, or other methods currently under development, expand our understanding of the disease’s status and serve as essential prognostic markers.

Notes

Conflicts of Interest

None declared.

REFERENCES

1. Kumar S, Paiva B, Anderson KC, Durie B, Landgren O, Moreau P, et al. 2016; International Myeloma Working Group consensus criteria for response and minimal residual disease assessment in multiple myeloma. Lancet Oncol. 17:e328–46. DOI: 10.1016/S1470-2045(16)30206-6. PMID: 27511158.
2. Paiva B, van Dongen JJ, Orfao A. 2015; New criteria for response assessment: role of minimal residual disease in multiple myeloma. Blood. 125:3059–68. DOI: 10.1182/blood-2014-11-568907. PMID: 25838346. PMCID: PMC4513329.
3. Paiva B, Gutiérrez NC, Rosiñol L, Vídriales MB, Montalbán MÁ, Martínez-López J, et al. 2012; 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. 119:687–91. DOI: 10.1182/blood-2011-07-370460. PMID: 22128143.
4. Yee AJ, Raje N. 2021; Minimal residual disease in multiple myeloma: why, when, where. Hematology Am Soc Hematol Educ Program. 2021:37–45. DOI: 10.1182/hematology.2021000230. PMID: 34889430. PMCID: PMC8791109.
5. Munshi NC, Avet-Loiseau H, Anderson KC, Neri P, Paiva B, Samur M, et al. 2020; A large meta-analysis establishes the role of MRD negativity in long-term survival outcomes in patients with multiple myeloma. Blood Adv. 4:5988–99. DOI: 10.1182/bloodadvances.2020002827. PMID: 33284948. PMCID: PMC7724898.
6. San Miguel JF, Almeida J, Mateo G, Bladé J, López-Berges C, Caballero D, et al. 2002; Immunophenotypic evaluation of the plasma cell compartment in multiple myeloma: a tool for comparing the efficacy of different treatment strategies and predicting outcome. Blood. 99:1853–6. DOI: 10.1182/blood.V99.5.1853. PMID: 11861305.
7. Puig N, Sarasquete ME, Balanzategui A, Martínez J, Paiva B, García H, et al. 2014; Critical evaluation of ASO RQ-PCR for minimal residual disease evaluation in multiple myeloma. A comparative analysis with flow cytometry. Leukemia. 28:391–7. DOI: 10.1038/leu.2013.217. PMID: 23860448.
8. Bakkus MH, Bouko Y, Samson D, Apperley JF, Thielemans K, Van Camp B, et al. 2004; Post-transplantation tumour load in bone marrow, as assessed by quantitative ASO-PCR, is a prognostic parameter in multiple myeloma. Br J Haematol. 126:665–74. DOI: 10.1111/j.1365-2141.2004.05120.x. PMID: 15327517.
9. Flores-Montero J, Sanoja-Flores L, Paiva B, Puig N, García-Sánchez O, Böttcher S, et al. 2017; Next Generation Flow for highly sensitive and standardized detection of minimal residual disease in multiple myeloma. Leukemia. 31:2094–103. DOI: 10.1038/leu.2017.29. PMID: 28104919. PMCID: PMC5629369.
10. Ladetto M, Brüggemann M, Monitillo L, Ferrero S, Pepin F, Drandi D, et al. 2014; Next-generation sequencing and real-time quantitative PCR for minimal residual disease detection in B-cell disorders. Leukemia. 28:1299–307. DOI: 10.1038/leu.2013.375. PMID: 24342950.
11. Takamatsu H. 2017; Comparison of minimal residual disease detection by multiparameter flow cytometry, ASO-qPCR, droplet digital PCR, and deep sequencing in patients with multiple myeloma who underwent autologous stem cell transplantation. J Clin Med. 6:91. DOI: 10.3390/jcm6100091. PMID: 28946710. PMCID: PMC5664006. PMID: 7d64e3ad7d5845fca141c797f00f606d.
12. Costa LJ, Derman BA, Bal S, Sidana S, Chhabra S, Silbermann R, et al. 2021; International harmonization in performing and reporting minimal residual disease assessment in multiple myeloma trials. Leukemia. 35:18–30. DOI: 10.1038/s41375-020-01012-4. PMID: 32778736.
13. Maclachlan KH, Came N, Diamond B, Roshal M, Ho C, Thoren K, et al. 2021; Minimal residual disease in multiple myeloma: defining the role of next generation sequencing and flow cytometry in routine diagnostic use. Pathology. 53:385–99. DOI: 10.1016/j.pathol.2021.02.003. PMID: 33674146.
14. Roshal M, Flores-Montero JA, Gao Q, Koeber M, Wardrope J, Durie BGM, et al. 2017; MRD detection in multiple myeloma: comparison between MSKCC 10-color single-tube and EuroFlow 8-color 2-tube methods. Blood Adv. 1:728–32. DOI: 10.1182/bloodadvances.2016003715. PMID: 29296716. PMCID: PMC5728052.
15. Blum A, Haussmann K, Streitz M, Schlickeiser S, Tietze-Buerger C, Blau IW, et al. 2019; Standardized assay for assessment of minimal residual disease in blood, bone marrow and apheresis from patients with plasma cell myeloma. Sci Rep. 9:2922. DOI: 10.1038/s41598-019-39631-2. PMID: 30814612. PMCID: PMC6393516.
16. Bayly E, Nguyen V, Binek A, Piggin A, Baldwin K, Westerman D, et al. 2020; Validation of a modified pre-lysis sample preparation technique for flow cytometric minimal residual disease assessment in multiple myeloma, chronic lymphocytic leukemia, and B-non Hodgkin lymphoma. Cytometry B Clin Cytom. 98:385–98. DOI: 10.1002/cyto.b.21893. PMID: 32530574.
17. Soh KT, Came N, Otteson GE, Jevremovic D, Shi M, Olteanu H, et al. 2022; Evaluation of multiple myeloma measurable residual disease by high sensitivity flow cytometry: an international harmonized approach for data analysis. Cytometry B Clin Cytom. 102:88–106. DOI: 10.1002/cyto.b.22053. PMID: 35005838. PMCID: PMC10105368.
18. Muz B, de la Puente P, Azab F, Luderer MJ, King J, Vij R, et al. 2016; A CD138-independent strategy to detect minimal residual disease and circulating tumour cells in multiple myeloma. Br J Haematol. 173:70–81. DOI: 10.1111/bjh.13927. PMID: 26729247. PMCID: PMC4918510.
19. Oberle A, Brandt A, Alawi M, Langebrake C, Janjetovic S, Wolschke C, et al. 2017; Long-term CD38 saturation by daratumumab interferes with diagnostic myeloma cell detection. Haematologica. 102:e368–70. DOI: 10.3324/haematol.2017.169235. PMID: 28522580. PMCID: PMC5685239. PMID: 5c70616cb8f34e96af6fe21af717ee6a.
20. Courville EL, Yohe S, Shivers P, Linden MA. 2020; VS38 identifies myeloma cells with dim CD38 expression and plasma cells following daratumumab therapy, which interferes with CD38 detection for 4 to 6 months. Am J Clin Pathol. 153:221–8. DOI: 10.1093/ajcp/aqz153. PMID: 31679012.
21. Rai S, Das N, Gupta R, Kumar L, Sharma A, Singh S, et al. 2023; Utility of CD229 as novel marker in measurable residual disease assessment in multiple myeloma-An evidence-based approach. Int J Lab Hematol. 45:179–86. DOI: 10.1111/ijlh.13992. PMID: 36403963.
22. Soh KT, Tario JD Jr. 2021; , Hahn T, Hillengass J, McCarthy PL, Wallace PK. CD319 (SLAMF7) an alternative marker for detecting plasma cells in the presence of daratumumab or elotuzumab. Cytometry B Clin Cytom. 100:497–508. DOI: 10.1002/cyto.b.21961. PMID: 33017079. PMCID: PMC8940539.
23. Ching T, Duncan ME, Newman-Eerkes T, McWhorter MME, Tracy JM, Steen MS, et al. 2020; Analytical evaluation of the clonoSEQ Assay for establishing measurable (minimal) residual disease in acute lymphoblastic leukemia, chronic lymphocytic leukemia, and multiple myeloma. BMC Cancer. 20:612. DOI: 10.1186/s12885-020-07077-9. PMID: 32605647. PMCID: PMC7325652. PMID: f4765b6429fd4f42acca9201ac81c886.
24. Dunn-Walters D, Townsend C, Sinclair E, Stewart A. 2018; Immunoglobulin gene analysis as a tool for investigating human immune responses. Immunol Rev. 284:132–47. DOI: 10.1111/imr.12659. PMID: 29944755. PMCID: PMC6033188.
25. Kim M, Jeon K, Hutt K, Zlotnicki AM, Kim HJ, Lee J, et al. 2021; Immunoglobulin gene rearrangement in Koreans with multiple myeloma: clonality assessment and repertoire analysis using next-generation sequencing. PLoS One. 16:e0253541. DOI: 10.1371/journal.pone.0253541. PMID: 34166440. PMCID: PMC8224885. PMID: 80888204ee10431d83c540911ac93584.
26. Romano A, Palumbo GA, Parrinello NL, Conticello C, Martello M, Terragna C. 2019; Minimal residual disease assessment within the bone marrow of multiple myeloma: a review of caveats, clinical significance and future perspectives. Front Oncol. 9:699. DOI: 10.3389/fonc.2019.00699. PMID: 31482061. PMCID: PMC6710454. PMID: c14d70f7b0e7430e93311b804b9d8e01.
27. Ng DP. 2021; Flow cytometric myeloma measurable residual disease testing in the era of targeted therapies. Int J Lab Hematol. 43(S1):71–7. DOI: 10.1111/ijlh.13587. PMID: 34288444.
28. Arcila ME, Yu W, Syed M, Kim H, Maciag L, Yao J, et al. 2019; Establishment of immunoglobulin heavy (IGH) chain clonality testing by next-generation sequencing for routine characterization of B-cell and plasma cell neoplasms. J Mol Diagn. 21:330–42. DOI: 10.1016/j.jmoldx.2018.10.008. PMID: 30590126. PMCID: PMC6436112.
29. Ho C, Syed M, Roshal M, Petrova-Drus K, Moung C, Yao J, et al. 2021; Routine evaluation of minimal residual disease in myeloma using next-generation sequencing clonality testing: feasibility, challenges, and direct comparison with high-sensitivity flow cytometry. J Mol Diagn. 23:181–99. DOI: 10.1016/j.jmoldx.2020.10.015. PMID: 33217553. PMCID: PMC7874334.
30. Ha J, Lee H, Shin S, Cho H, Chung H, Jang JE, et al. 2022; Ig gene clonality analysis using next-generation sequencing for improved minimal residual disease detection with significant prognostic value in multiple myeloma patients. J Mol Diagn. 24:48–56. DOI: 10.1016/j.jmoldx.2021.09.006. PMID: 34656761.
31. Ho C, Rothberg PG. 2021; Next-generation sequencing-based antigen-receptor gene clonality assays: will they become the clinical standard? J Mol Diagn. 23:1043–6. DOI: 10.1016/j.jmoldx.2021.07.002. PMID: 34293488.
32. Brüggemann M, Kotrová M, Knecht H, Bartram J, Boudjogrha M, Bystry V, et al. 2019; Standardized next-generation sequencing of immunoglobulin and T-cell receptor gene recombinations for MRD marker identification in acute lymphoblastic leukaemia; a EuroClonality-NGS validation study. Leukemia. 33:2241–53. DOI: 10.1038/s41375-019-0496-7. PMID: 31243313. PMCID: PMC6756028.
33. Cavo M, San-Miguel J, Usmani SZ, Weisel K, Dimopoulos MA, Avet-Loiseau H, et al. 2022; Prognostic value of minimal residual disease negativity in myeloma: combined analysis of POLLUX, CASTOR, ALCYONE, and MAIA. Blood. 139:835–44. DOI: 10.1182/blood.2021011101. PMID: 34289038. PMCID: PMC8832474.
34. Martinez-Lopez J, Lahuerta JJ, Pepin F, González M, Barrio S, Ayala R, et al. 2014; Prognostic value of deep sequencing method for minimal residual disease detection in multiple myeloma. Blood. 123:3073–9. DOI: 10.1182/blood-2014-01-550020. PMID: 24646471. PMCID: PMC4023416.
35. Avet-Loiseau H, Lauwers-Cances V, Corre J, Moreau P, Attal M, Munshi N. 2017; Minimal residual disease in multiple myeloma: final analysis of the IFM2009 trial. Blood. 130(S1):435.
36. Yao Q, Bai Y, Orfao A, Chim CS. 2019; Standardized minimal residual disease detection by next-generation sequencing in multiple myeloma. Front Oncol. 9:449. DOI: 10.3389/fonc.2019.00449. PMID: 31245284. PMCID: PMC6563351. PMID: 95a5e973153148678046ecda34f8b8d5.
37. Rustad EH, Hultcrantz M, Yellapantula VD, Akhlaghi T, Ho C, Arcila ME, et al. 2019; Baseline identification of clonal V(D)J sequences for DNA-based minimal residual disease detection in multiple myeloma. PLoS One. 14:e0211600. DOI: 10.1371/journal.pone.0211600. PMID: 30901326. PMCID: PMC6430394. PMID: a68a364060d44da6965935cdc49fd95f.
38. Medina A, Jiménez C, Sarasquete ME, González M, Chillón MC, Balanzategui A, et al. 2020; Molecular profiling of immunoglobulin heavy-chain gene rearrangements unveils new potential prognostic markers for multiple myeloma patients. Blood Cancer J. 10:14. DOI: 10.1038/s41408-020-0283-8. PMID: 32029700. PMCID: PMC7004993.
39. Medina A, Puig N, Flores-Montero J, Jimenez C, Sarasquete ME, Garcia-Alvarez M, et al. 2020; Comparison of next-generation sequencing (NGS) and next-generation flow (NGF) for minimal residual disease (MRD) assessment in multiple myeloma. Blood Cancer J. 10:108. DOI: 10.1038/s41408-020-00377-0. PMID: 33127891. PMCID: PMC7603393.
40. Martinez-Lopez J, Lahuerta JJ, Pepin F, González M, Barrio S, Ayala R, et al. 2014; Prognostic value of deep sequencing method for minimal residual disease detection in multiple myeloma. Blood. 123:3073–9. DOI: 10.1182/blood-2014-01-550020. PMID: 24646471. PMCID: PMC4023416.
41. Oliva S, Genuardi E, Belotti A, Frascione PMM, Galli M, Capra A, et al. 2020; Multiparameter flow cytometry (MFC) and next generation sequencing (NGS) for minimal residual disease (MRD) evaluation: results of the FORTE trial in newly diagnosed multiple myeloma (MM). J Clin Oncol. 38(S):8533. DOI: 10.1200/JCO.2020.38.15_suppl.8533.
42. Avet-Loiseau H, Bene MC, Wuilleme S, Corre J, Attal M, Arnulf B, et al. 2019; Concordance of post-consolidation minimal residual disease rates by multiparametric flow cytometry and next-generation sequencing in CASSIOPEIA. Clinical Lymphoma Myeloma and Leukemia. 19(S):e3–4. DOI: 10.1016/j.clml.2019.09.005.
43. Cho H, Shin S, Chung H, Jang JE, Kim YR, Cheong JW, et al. 2022; Real-world data on prognostic value of measurable residual disease assessment by fragment analysis or next-generation sequencing in multiple myeloma. Br J Haematol. 198:503–14. DOI: 10.1111/bjh.18211. PMID: 35505579.
44. Ferla V, Antonini E, Perini T, Farina F, Masottini S, Malato S, et al. 2022; Minimal residual disease detection by next-generation sequencing in multiple myeloma: promise and challenges for response-adapted therapy. Front Oncol. 12:932852. DOI: 10.3389/fonc.2022.932852. PMID: 36052251. PMCID: PMC9426755. PMID: 3a4a5cbe7c4e469dae9619fa211f2045.
45. Charalampous C, Kourelis T. 2021; Minimal residual disease assessment in multiple myeloma patients: minimal disease with maximal implications. Front Oncol. 11:801851. DOI: 10.3389/fonc.2021.801851. PMID: 35155198. PMCID: PMC8825476. PMID: d668e33adf5d4a09b2466986a6086063.
46. Garcés JJ, San-Miguel J, Paiva B. 2022; Biological characterization and clinical relevance of circulating tumor cells: opening the Pandora's box of multiple myeloma. Cancers (Basel). 14:1430. DOI: 10.3390/cancers14061430. PMID: 35326579. PMCID: PMC8946760. PMID: 635d676d767a479aaae7cfdf5edf3b13.
47. Sanoja-Flores L, Flores-Montero J, Garcés JJ, Paiva B, Puig N, García-Mateo A, et al. 2018; Next generation flow for minimally-invasive blood characterization of MGUS and multiple myeloma at diagnosis based on circulating tumor plasma cells (CTPC). Blood Cancer J. 8:117. DOI: 10.1038/s41408-018-0153-9. PMID: 30455467. PMCID: PMC6242818.
48. Sanoja-Flores L, Flores-Montero J, Pérez-Andrés M, Puig N, Orfao A. 2020; Detection of circulating tumor plasma cells in monoclonal gammopathies: methods, pathogenic role, and clinical implications. Cancers (Basel). 12:1499. DOI: 10.3390/cancers12061499. PMID: 32521788. PMCID: PMC7352573. PMID: c22d6affe7894a69969c1d56dd2cd897.
49. Gonsalves WI, Rajkumar SV, Dispenzieri A, Dingli D, Timm MM, Morice WG, et al. 2017; Quantification of circulating clonal plasma cells via multiparametric flow cytometry identifies patients with smoldering multiple myeloma at high risk of progression. Leukemia. 31:130–5. DOI: 10.1038/leu.2016.205. PMID: 27457702. PMCID: PMC5244483.
50. Periago A, Campillo JA, Mrowiec A, Gimeno L, Montes NR, Martínez-Sánchez MV, et al. 2016; Circulating aberrant plasma cells allows risk stratification of patients with myeloma. Am J Hematol. 91:E353–5. DOI: 10.1002/ajh.24431. PMID: 27220360.

Table 1
Comparison of NGF and NGS-based MRD assays
NGF-based MRD NGS-based MRD
Applicability (%) Nearly 100% >90%
Sensitivity 10-5–10-6 Up to 10-6
Sample type Cells Cells, DNA
Cell acquisition 10 million cells needed 2–3 million cells needed
Sample processing Fresh samples Can use both fresh and stored samples
Baseline sample testing Not mandatory Mandatory
Turnaround time 4 h Several days
Standardization EuroFlow consortium Commercial companies
Availability Widely available, in-house or commercial kits US FDA-approved platform not available in Korea
Other commercial kits or in-house sequencing possible
Cost +/++ ++/+++

Abbreviations: NGF, next-generation flow; NGS, next-generation sequencing; US, United States; FDA, Food and Drug Administration.

TOOLS
Similar articles