Skip to main content
Erschienen in: Discover Oncology 1/2024

Open Access 01.12.2024 | Research

Evaluation of next-generation sequencing versus next-generation flow cytometry for minimal-residual-disease detection in Chinese patients with multiple myeloma

verfasst von: Mo Zhou, Yan Chen, Yanlei Gong, Mingqing Zhu, Jiannong Cen, Jinlan Pan, Lingzhi Yan, Jingjing Shang, Song Jin, Xiaolan Shi, Weiqin Yao, Shuang Yan, Depei Wu, Suning Chen, Chengcheng Fu, Li Yao

Erschienen in: Discover Oncology | Ausgabe 1/2024

Abstract

Purpose

To evaluate the efficacy of next-generation sequencing (NGS) in minimal-residual-disease (MRD) monitoring in Chinese patients with multiple myeloma (MM).

Methods

This study analyzed 60 Chinese MM patients. During MRD monitoring in these patients’ post-therapy, clonal immunoglobulin heavy chain (IGH) rearrangements were detected via NGS using LymphoTrack assays. MRD monitoring was performed using NGS or next-generation flow cytometry (NGF), and the results were compared. Additionally, the sensitivity and reproducibility of the NGS method were assessed.

Results

The MRD detection range of the NGS method was 10–6–10–1, which suggested good linearity, with a Pearson correlation coefficient of 0.985 and a limit of detection of 10–6. Intra- and inter-assay reproducibility analyses showed that NGS exhibited 100% reproducibility with low variability in clonal cells. At diagnosis, unique clones were found in 42 patients (70.0%) with clonal IGH rearrangements, which were used as clonality markers for MRD monitoring post-therapy. Comparison of NGS and NGF for MRD monitoring showed 79.1% concordance. No samples that tested MRD-positive via NGF were found negative via NGS, indicating the higher sensitivity of NGS. MRD could be detected using NGS in 6 of 7 samples before autologous hematopoietic stem-cell transplantation, and 5 of them tested negative post-transplantation. In contrast, the NGF method could detect MRD in only 1 sample pre-transplantation.

Conclusion

Compared with NGF, NGS exhibits higher sensitivity and reproducibility in MRD detection and can be an effective strategy for MRD monitoring in Chinese MM patients.
Hinweise

Supplementary Information

The online version contains supplementary material available at https://​doi.​org/​10.​1007/​s12672-024-00938-w.
Mo Zhou and Yan Chen contributed equally to this work.

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

1 Introduction

Multiple myeloma (MM) is a clonal plasma-cell tumor, with high cytogenetic heterogeneity [1]. It is the second most common hematological malignancy in many countries [24] and has a substantial impact on public health. The specific incidence of MM in China is not clear, but the median age of onset is 57.9–59 years old [5, 6], which is far lower than that observed in western countries [79]. In recent years, new chemotherapies and immune drugs have greatly improved the remission rate and survival time in MM [4]. However, as the long-term survival rate of patients has increased, the recurrence rate has also risen, resulting in eventual relapse for the majority of patients, for whom there is currently no cure [10]. Currently, the treatment goals in MM prioritize long-term disease control, disease remission, and improvement of the life quality of the patient. Consequently, there is an urgent need for improved curative outcomes. Additionally, there is an urgent need for sensitive methods for continuous monitoring of minimal residual disease (MRD) and guiding clinical treatment following complete remission [11].
MM represents a group of malignant tumors originating from B lymphocytes. During the normal formation of B cells and their differentiation into plasma cells, recombination of variable (V), diversity (D), and joining (J) gene segments leads to immunoglobulin heavy chain (IGH) rearrangements. The diversity, randomness, and arbitrary insertion and deletion of nucleotides in the junction region during IGH rearrangements result in each B-cell clone having a unique VDJ sequence resulting from its IGH rearrangement [12]. Therefore, the clonal IGH rearrangement identified at the time of diagnosis can serve as a valuable clonal biomarker for MRD monitoring during clinical treatment.
The capillary electrophoresis (CE) method, which has gradually become a reference standard for detecting gene rearrangements, particularly for detecting clonal IGH rearrangements, can be used as an auxiliary diagnostic tool for lymphoid tumors. Yet, its application for MRD monitoring is limited [12]. In contrast, the next-generation sequencing (NGS) technology offers multiple advantages, such as being high-throughput as well as highly time-effective, accurate, and informative. These qualities make NGS highly beneficial in accurately detecting and quantifying clonal rearrangements, especially in the context of MRD monitoring in MM patients post-treatment [13].
The International Myeloma Working Group (IMWG) recommends next-generation flow cytometry (NGF) and NGS as the main methods for evaluating MRD [14]. However, the sensitivity, repeatability, and standardization of clinical correlation analysis of these technologies are still under exploration [15, 16]. In addition, NGS is rarely used to detect MRD in MM patients in China. Thus, this study aimed to assess the applicability of NGS versus NGF to detecting clonal IGH rearrangements in newly diagnosed MM patients and to MRD monitoring post-treatment.

2 Methods

2.1 Patients

This retrospective study included 60 newly diagnosed MM patients from the First Affiliated Hospital of Soochow University between September 2019 and October 2020. All the samples from these patients were collected from the biological sample bank used in a previous study [17], during which the patients were treated with bortezomib, lenalidomide, and dexamethasone with or without subsequent autologous stem-cell transplantation (ASCT).
This study was approved by the Ethics Committee of the First Affiliated Hospital of Soochow University and followed the Declaration of Helsinki. Informed consent was obtained from each patient.

2.2 Cytogenetic analysis

At diagnosis, interphase fluorescence in situ hybridization (FISH) analysis was performed alongside sorting CD138 plasma cells. The following probes were used: 1q21 for 1q gain, RB1 and D13S319 for 13q deletion or monosomy 13, TP53 for TP53 deletion or monosomy 17, IGH for 14q32 rearrangements, and probes targeting the individual IGH rearrangements t (11;14)(q13; q32) CCND::IGH, t (4;14)(p16; q32) FGFR3::IGH, and t (14;16)(q32; q23) IGH::MAF. The probes were obtained from GP Medical Technologies, Beijing, China. A minimum of 200 interphase nuclei obtained from bone-marrow (BM) cultures were analyzed using a Leica DMRXA fluorescence microscope (Leica, Wetzlar, Germany).

2.3 DNA extraction

Genomic DNA was isolated from BM aspirates at the time of diagnosis and the follow-up period by using a DNA Extraction Kit (Promega, USA). DNA quantity and quality were evaluated using a Qubit instrument (Thermo Fisher, USA) according to the manufacturer’s instructions.

2.4 PCR and CE

PCR and CE were used to detect IGH clonality [18]. In brief, the IGH-FR1, FR2, and FR3 gene rearrangements were amplified using PCR with the IdentiCloneTM rearrangement detection kit (Invivoscribe, USA) according to the manufacturer’s instructions. An ABI3730 gene analyzer (Thermo Fisher, USA) was used to analyze the amplicons, following previously described scoring criteria [19].

2.5 NGF

The flow-cytometry-based strategy used in this study involved 10 characteristic antibodies, including 8 against cell-membrane proteins (CD138-APC, CD38-APC750, CD19-ECD, CD45-KO, CD56-PC7, CD27-PB, CD81-APC700, and CD117-PC5; from Beckman Coulter, USA), and 2 against cytoplasmic proteins (Kappa-FITC and Lambda-PE; from Dako, Denmark), which have previously been described to be able to differentiate between abnormal and normal plasma cells via flow cytometry [2022]. Sample preparation and detection were performed as previously described [17]. Briefly, 200 μL–5 mL BM aspirates [(2–20) × 106 cells] were stained with the monoclonal antibodies for 30 min at 20–30 °C after lysing red-blood cells by using ammonium chloride and then washed with phosphate-buffered saline. For intracellular light-chain evaluation, cells were stained with anti-kappa and anti-lambda antibodies after adding membrane breakers, followed by washing and incubation. Flow-cytometry events were acquired and analyzed using a Navios Flow cytometer (Beckman Coulter, USA). One million nucleated cells were obtained each time, and if ≥ 20 abnormal plasma cells were detected, the result was considered positive.

2.6 Clonality testing via NGS

NGS-based clonality testing was performed using commercially available LymphoTrack assays (Invivoscribe, USA) targeting IGH-FR1, IGH-FR2, and IGH-FR3. The assays were performed following the manufacturer’s instructions. PCR amplification was performed using a master mix containing primers that had barcoded sequence adaptors. After the PCR products were purified and quantified, they were sequenced using an Ion S5 sequencer platform (Thermo Fisher, USA). Sequencing data in FASTQ format were analyzed using the LymphoTrack software package (InVivoScribe Technologies, San Diego, CA) [16, 23].

2.7 Criteria for clonality and MRD

The criteria for determining the IGH clonality in newly diagnosed MM patients were as follows: a minimum of five identical sequences obtained through sequencing constituted a clone, and the frequency of the clone needed to be > 5% to be used as a marker for MRD tracking in MM patients, as previously described [13]. All the sequences identified in BM samples from patients in the remission stage were compared with the clonal sequences derived from the tumor cells in the newly diagnosed samples, serving as index clonal sequences. Samples were considered MRD-positive if the same sequences were detected [24].

2.8 Validation of the NGS-based method

To assess the performance characteristics of the NGS-based method, a series of experiments were performed. Commercially available assays containing a positive clonal control (IVS0019) and a polyclonal negative control (IVS0000) were used to prepare successive dilutions of DNA, spanning the range of 10–6–10–1, to determine the limit of detection of the method. To assess the specificity of this method, patients exhibiting clonality were identified, providing specific clone information. Three samples with different MRD levels were selected. Accordingly, corresponding patients P1, P2, and P3 had MRD levels of 10–2, 10–3, and 10–4, respectively. The NGS results of these three samples were detected before with definite results, and the selection criteria were to cover the test range of different orders of magnitude of MRD. These samples were used for assessing inter- and intra-assay reproducibility.

2.9 Statistical analysis

All the data were statistically analyzed using GraphPad Prism version 9.3.1 (https://​www.​graphpad.​com/​scientific-software/​prism/​). Mean values were compared using the independent sample t-test and analysis of variance, and rates were compared using the χ2 test. Fisher’s exact test was used to test categorical variables, and the Spearman correlation coefficient was used for statistical comparison. A p-value < 0.05 was considered to indicate statistical significance. The graphs were generated using GraphPad Prism version 9.3.1 and R version 4.1.0 (R Core Team).

3 Results

3.1 Clinical characteristics of the patients

A total of 60 patients, consisting of 33 males and 27 females, with a median age of 57 (39–70) years, were enrolled in this study. The diagnosis of MM was based on the standard criteria established by the IMWG [14]. Among the patients, there were 28 IgG, 12 IgA, 5 IgD, 13 light-chain, and 2 non-secretory myeloma cases. The cytogenetic abnormalities were detected in 40 cases and included t (4;14), t (11;14), t (14;16), 1q21 gain, and 17p abnormalities. Some cases had 2–3 simultaneous abnormalities. Cytogenetic risk stratification was performed according to the Revised International Staging System (R-ISS) [25]. Accordingly, the R-ISS score was 1 in 9 patients, 2 in 39 patients, and 3 in 8 patients. Additionally, 55 patients were at Durie–Salmon stage III at the time of diagnosis. Among the 60 patients, samples were collected from 36 at initial diagnosis and after remission post-induction therapy, with an additional 7 patients undergoing ASCT transplantation. The clinical characteristics of the participants are presented in Table 1.
Table 1
Characteristics of the patients
Characteristics
Patients
Gender, male/female
60 (33/27)
Median age, years (range)
56 (39–70)
M protein type
 IgH
  IgG
28
  IgA
12
  IgD
5
  Non-secretory
2
 IgL
13
  Kappa chain
1
  Lambda chain
12
Cytogenetics
 t (4;14)
18
 t (11;14)
10
 t (14;16)
1
 1q21 gain
33
 17p abnormalities
7
Cytogenetic risk
 High risk
26
 Standard risk
34
DS
 Stage I, II
5 (8.33%)
 Stage III
55 (91.67%)
R-ISSa
 Stage I
9 (15%)
 Stage II
23 (38.33%)
 Stage III
28 (46.67%)
Hemoglobin (g/L, \(\bar{\text{x}} \pm {\text{s}}\))
93 ± 27
Albumin (g/L, \(\bar{\text{x}} \pm {\text{s}}\))
34.1 ± 7.8
LDH, µmol/L (range)
180.8 (93.3–542.1)
Creatinine, µmol/L (range)
129.07 (39.1–650.5)
Calcium, µmol/L (range)
2.35 (1.94–3.63)
IgH immunoglobulin heavy chain, IgL immunoglobulin light chain, DS Durie–Salmon, R-ISS revised international staging system, LDH lactate dehydrogenase

3.2 Clonality detection by using NGS

All the 60 patients were analyzed using both NGS and CE at the time of diagnosis and tested for FR1, FR2, and FR3 regions simultaneously. The consistency rates for IGH FR1, IGH FR2, and IGH FR3 were 91.7% (55/60), 93.3% (56/60), and 96.7% (58/60), respectively (Fig. 1a). The overall concordance between the two methods was 98.3% (59/60). As shown in Fig. 1b, clonal IGH-FR1, IGH-FR2, and IGH-FR3 rearrangements were detected in 37, 38, and 29 cases via the NGS-based method, respectively, with detection rates of 61.7% (37/60), 63.3% (38/60), and 48.3% (29/60). The overall detection rate of the NGS method was 70.0% (42/60) when considering the combined detection of IGH-FR1/FR2/FR3 rearrangements. The details were described in our previous publication [26].
As shown in Table 2, patients with clonal IGH rearrangements and those with polyclonal rearrangements were compared in terms of the immunoglobulin type, cytogenetic risk stratification, chemotherapy regimen, and ASCT. There was a positive correlation between clonal IGH rearrangement and heavy-chain myeloma cases, including IgG, IgA, IgD, and non-secretory types. No significant difference in distribution was observed for other factors, such as cytogenetics and chemotherapy regimens. Notably, the detection rate of clonal IGH rearrangement significantly differed between the heavy-chain and light-chain myeloma cases (P = 0.0012).
Table 2
Comparison between the patients carrying clonal IGH rearrangements and those carrying polyclonal rearrangements
Characteristics
Total cases
Clonala
Polyclonala
P-value
Ig
0.0012
 IgH
47 (78.33%)
38 (63.33%)
9 (15%)
 
 IgL
13 (21.67%)
4 (6.67%)
9 (15%)
 
Cytogeneticsb
0.3979
 SR cytogenetics
34 (56.67%)
22 (36.67%)
12 (20%)
 
 HR cytogenetics
26 (43.33%)
20 (33.33%)
6 (10%)
 
VRd treatment
0.2551
 Yes
51 (85%)
34 (56.67%)
17 (28.33%)
 
 No
9 (15%)
8 (13.33%)
1 (11.11%)
 
Transplant
0.7080
 Yes
50 (83.33%)
34 (56.67%)
16 (26.67%)
 
 No
10 (16.67%)
8 (13.33%)
2 (3.33%)
 
IgH immunoglobulin heavy chain, IgL immunoglobulin light chain, HR cytogenetics high-risk cytogenetics, SR cytogenetics standard-risk cytogenetics, VRd bortezomib, lenalidomide and dexamethasone
aClonality of IGH gene rearrangement was determined by the NGS method
bHR cytogenetics were defined as the presence of t (4;14)(p16; q32), t (14;16)(q32; q23), and /or 17p abnormalities. SR cytogenetics: cytogenetic/FISH test done and high risk not explicitly mentioned

3.3 Specificity of NGS

The CDR3 sequence detected using NGS was unique in each patient. As shown in Table 3, among the 42 patients with clonal IGH rearrangements (P1–P42) detected using NGS, each had their own unique IGH rearrangement. Additionally, in 3 patients, bi-allelic rearrangement clones, consisting of one productive and one unproductive IGH rearrangement, were detected. Productive IGH rearrangements were detectable in all the 42 newly diagnosed patients. Analysis of the complementarity-determining region 3 (CDR3) of each IGH rearrangement revealed that the CDR3 sequence of each patient was unique and comprised different V, D, and J gene fragments. The median length of the CDR3 region was 48 base pairs (bp), with a range of 27–78 bp, and the median clonal frequency was 22.7% (5.44–88.26%). As expected, polyclonal IGH rearrangements were detected in peripheral blood samples from healthy individuals.
Table 3
Cloning features of IGH rearrangement with MM patients
Patient ID
Clonality
CDR3 nucleotide seq
Length (bp)
Productive
V gene
D gene
J gene
Frequency (%)
P1
clone1
GCGAGAGCCGGGCAAGAAGTTGCTTCTCGTTCCTACTACATGGACGTC
48
Yes
IGHV3-33
IGHD2-21
IGHJ6
22.87
P2
clone1
ACAAAAGGGGGTCGATATGGGCAGTGGCGGCGATTTGACTAC
42
Yes
IGHV3-23
IGHD6-19
IGHJ4
30.94
P3
clone1
ACCACAGATAGTAGCTCTCTCTACTAC
27
Yes
IGHV3-15
IGHD6-6
IGHJ4
24.50
P4
clone1
ATTTTTTCGACCGGCGGCGTCCGCTATAACGGTATGGACGTC
42
Yes
IGHV4-39
IGHD3-3
IGHJ6
25.92
P5
clone1
GCAAAAGACTTCTCTTTTCGATATAGCAGCAGCTGGTTCGGGGCATTTGACTCC
54
Yes
IGHV3-43
IGHD6-13
IGHJ4
35.13
P6
clone1
GCACACAGAGGTACGGTAGGAGTAGACCCTGTAGGGGGGTACTTCTACGCTATGGACGTC
60
Yes
IGHV2-5
IGHD4-17
IGHJ6
5.00
P7
clone1
GCACGCCTAGGATATTGCAGTAGTCTCAGTTGTTATCTTGACTAC
45
Yes
IGHV2-5
IGHD2-2
IGHJ4
26.89
P8
clone1
GCACGGATACGTGGAACTGGAGTTAGGGGCTATTACTACAGCTACTATGGTATGGACGTC
60
Yes
IGHV2-70
IGHD1-1
IGHJ6
33.86
P9
clone1
GCATTGAGTAACTGGCGCTTCTTCTTCGACTAC
33
Yes
IGHV1-3
IGHD1-20
IGHJ4
63.78
P10
clone1
GCCAGATTGGCGCCTACGGGCAACTGGTACTTCGACCTC
39
Yes
IGHV1-69
IGHD4-17
IGHJ2
88.26
P11
clone1
GCGAAAGCCTTCCCGCACGAATTTTTATGGGTGTTCTGGTTGCTAGTCGGAGCTTCAGGTG
61
No
IGHV7-4-1
IGHD1-26
IGHJ1
23.94
 
clone2
GTTACAACAGTGGCTGCGGGCGGGCAGTTTGACTCC
36
Yes
IGHV5-51
IGHD6-19
IGHJ4
5.44
P12
clone1
GCGAAAGCGAGCGGATATTGTGATAGTATCAGCTGCCATTTCCTCTTTGACTAC
54
Yes
IGHV3-23
IGHD2-2
IGHJ4
51.67
P13
clone1
GCGAAGGGGACGGGTGCTTATGACCTG
27
Yes
IGHV3-23
IGHD3-10
IGHJ3
44.92
P14
clone1
GCGAGAATCGGGGCGATGGTAGGAACTACTGACCAC
36
Yes
IGHV3-48
IGHD1-7
IGHJ4
27.21
P15
clone1
GCGAGACAATATATGAGTTCGTTGAACTGGTTCAACCCC
39
Yes
IGHV4-39
IGHD3-16
IGHJ5
56.76
P16
clone1
GCGAGACACATTGTTATCATACGAGCCGGCATGACGAGTGTTTACTACTACCTGGACGTC
60
Yes
IGHV4-59
IGHD3-3
IGHJ6
65.22
P17
clone1
GCGAGACAGGGTGGGACGCCTACATCGGACTTCTACTACTACGGTTTGGACGTC
54
Yes
IGHV3-30
IGHD1-26
IGHJ6
40.32
 
clone2
GCGAGAGACGGAGTGAGGATCTGGTGACACTTACATTACAGCCGTAAGGGCGTC
54
No
IGHV4-39
IGHD2-21
IGHJ6
9.49
P18
clone1
GCGAGACATGGTTATTACTTTGACAATACTGCTACGTTTGACTAT
45
Yes
IGHV4-39
IGHD3-22
IGHJ4
45.73
P19
clone1
GCGAGACCCCCACCCACGGTCTCTCGAGACTGGTATTTCGATCTC
45
Yes
IGHV3-7
IGHD4-17
IGHJ2
15.53
P20
clone1
GCGAGAGAATATCGGGCCACAGCTGGCGCAGCCTACTCCTTCTACGGTATGGACGTC
57
Yes
IGHV3-33
IGHD6-13
IGHJ6
69.96
P21
clone1
TCCACTGTTTGGATGTCCGACATAGAAGGCACGATTACCCGACGTGACCTC
51
Yes
IGHV3-15
IGHD5-12
IGHJ1
77.58
P22
clone1
GCGAGAGACTTGCTTCCGGGCGAGAGGTTCGGGGAGTGGCCCCCCACCTCCTATCACTACTACTACGGTATGGACGTC
78
Yes
IGHV1-8
IGHD3-10
IGHJ6
60.16
P23
clone1
GCGAGAGAGTTCTGGTACTTTGGGAGTTATTCTCCGCACTACTTCTACGGTATGGACATC
60
Yes
IGHV1-2
IGHD3-10
IGHJ6
43.00
P24
clone1
GCGAGAGATCGGGCCCCGGAAATTCGCGGAGTTCTGATAATAAATGATGACTTT
54
Yes
IGHV1-18
IGHD3-10
IGHJ4
26.37
P25
clone1
GCGAGAGCGTCTCTATTATGGGGATATTGTAGTAGTAGCAGCTGCTCCCTGCCGACCCCTATGGACGTC
69
Yes
IGHV4-34
IGHD2-2
IGHJ6
69.70
P26
clone1
GCGAGAGGCCCGCCAGTACAATATTGTAGTATCACCAGTTGTTATTTGTACCACTTAGACCAC
63
Yes
IGHV3-30
IGHD2-2
IGHJ4
69.33
P27
clone1
GCGAGAGGCGGAGGGCGAAATTACTATTACTACTTCCACATGGACGTC
48
Yes
IGHV7-4-1
N/A
IGHJ6
63.88
P28
clone1
GCGAGAGGCTCGAGAGGATATCTTTTTGACGAGCCAAATTCTAGGCCTCTTATCTACTATTATATAGACGTC
72
yes
IGHV4-59
IGHD2-2
IGHJ6
37.34
P29
clone1
GCGAGAGGTCGCGGATACTGTGATGGCGGTTACTGCACCTCGCGGGCCCCCTACACTCTAGACGTC
66
Yes
IGHV3-11
IGHD2-21
IGHJ6
59.02
P30
clone1
GCGAGAGTGCGGGATTCTGAGTTCTACTTTTTTGACTTC
39
Yes
IGHV3-21
IGHD1-26
IGHJ4
78.01
P31
clone1
GCGAGAGTGTTCGATAGTAGTGGTCTCTATTTCATCGCTTTTGACTCC
48
Yes
IGHV3-30
IGHD3-22
IGHJ4
66.98
P32
clone1
GCGAGCTGTGGTACTCCCAGCTGCTTTCTCGCCGCCCACCAT
42
Yes
IGHV3-21
IGHD2-2
IGHJ4
7.21
P33
clone1
GCGAGGGGGGGGACTCAGTGGAACGGCTATCTTGACTCC
39
Yes
IGHV1-18
IGHD1-1
IGHJ4
64.85
P34
clone1
GCGAGTTCCGTAGGCCACTATAAGTATTGGAGTGGTTATTTGAATTAC
48
Yes
IGHV5-51
IGHD3-3
IGHJ4
60.00
P35
clone1
GCGCGACATTCGCGGATTTATAACTGGTTCGACCCC
36
Yes
IGHV4-59
IGHD3-3
IGHJ5
31.29
P36
clone1
GCGCGCGACGGGGGAGTTCATTATCCTAAATTCTCCCACCACGGAATGGACGTC
54
Yes
IGHV1-18
IGHD3-16
IGHJ6
35.80
P37
clone1
GCGCGGATCTCAAAACTAAGGGGATATAGACACCTGCGTAGAGACTCCCTTTGGTACTACTTCTTCGGTATGGACGTC
78
Yes
IGHV2-70
IGHD5-12
IGHJ6
18.55
 
clone2
TTTGCGACTGTGACTTTATGGTGGTGACAACTGACGCCCGTGTTGACTCG
50
No
IGHV1-46
IGHD2-21
IGHJ4
15.00
P38
clone1
GCGGTGCGCAGGATGGATACTAGTGGTTGGTATCGGGGTTTTGACTCC
48
Yes
IGHV5-51
IGHD3-22
IGHJ4
14.89
P39
clone1
GTGAAAGATTGGGGACCCTACGGTGACTCTACCCGTGGAGACGTC
45
Yes
IGHV3-23
IGHD4-17
IGHJ6
7.35
P40
clone1
GTGAGAGACAACATTGTGGCGGTGACAGCTTTTCACCTGGACAATAAGGACGATGCTTTTGAAATA
66
Yes
IGHV1-18
IGHD2-21
IGHJ3
22.35
P41
clone1
GTGAGAGATTACGCCTGGGCCTTCGACACC
30
Yes
IGHV3-48
IGHD3-16
IGHJ5
26.29
P42
clone1
GTGGACCCGGGATGGGGTCTTCAGTCCACTGTGGACGTC
39
Yes
IGHV3-30
IGHD3-16
IGHJ6
49.06

3.4 The limit and linearity of detection of NGS

To assess the sensitivity of NGS, serially diluted clonal lymphoid DNA was prepared. The IGH rearrangement clones with serial dilutions were detected as the results of MRD. As shown in Fig. 2a, the index clonal sequence was detectable at all the dilution levels and showed a good linear relationship. When the total input was increased to 3 μg DNA in three replicates (1 μg each), the index clonal sequence remained detectable down to 10–6 dilution.

3.5 Reproducibility of NGS

Three samples with different MRD levels, previously detected with definitive results, were selected to detect cells of interest at a 0.01% level. Each sample was tested in triplicate to evaluate inter- and intra-assay reproducibility. The results corresponding to the index clone sequence are shown in Fig. 2b. The results within each triplicate were highly consistent, with the index clone being detectable at 10–2, 10–3, and 10–4 dilutions. Clonal cells exhibited 100% reproducibility with low variability (6.13–10.85% coefficient of variation). These findings indicate good reproducibility of NGS in detecting MRD.

3.6 MRD monitoring of MM patients during the follow-up period

After verifying the performance of NGS in detecting clonal IGH rearrangements, MRD was monitored in 36 patients by using NGS in parallel to NGF. A total of 43 samples from these patients, collected at different time points during MRD monitoring, were tested using both NGS and NGF. Figure 3 shows the comparison of the results derived from the NGS-based analysis (X-axis, assessed at 10–6) and those from the NGF-based one (Y-axis, assessed at 10–5) in 43 samples from MM patients during the follow-up period. The overall consistency between the two methods was 34/43 (79.1%). The levels of MRD determined using NGS were highly correlated with those determined using NGF (Spearman coefficient R = 0.8047). Specifically, all the 16 samples identified as MRD-positive via NGF were also detected as positive via NGS. However, 9 samples that were detected as MRD-positive via NGS were negative according to NGF results. The remaining 18 samples tested MRD-negative via both methods.
Among these patients, 36 were followed up to the remission stage after induction therapy, and 7 of these 36 patients underwent ASCT. MRD detection was performed on samples from these 7 patients before and after ASCT. Details about these 7 patients are shown in Table 4. Among the 7 samples taken before ASCT, only 1 sample tested MRD-positive via NGF. In contrast, 6 of these 7 samples tested positive via NGS. Of these 6 patients, 4 tested negative via NGS after ASTC, and the remaining 2 patients were still positive, indicating a reduction in the number of MRD-positive patients relative to the number before ASTC. Notably, all the post-ASCT samples tested negative via NGF.
Table 4
MRD monitoring in MM patients post-ASCT
NO
Sex
Age
Type
DS
ISS
R-ISS
Cytogenetics
Induction regimen
After induction
After ASCT
Outcome
Response
NGF-MRD
NGS-MRD
NGF-MRD
NGS-MRD
P1
F
46y
IgA-κ
IIIA
II
II
t(4;14),1q21+
VRd
sCR
0
8.06E−05
0
0
Alive at 24mo
P2
F
46y
λ
IIIA
III
II
t(11;14),1q21+
VRd
sCR
0
7.39E−04
0
0
Alive at 34mo
P3
F
62y
λ
IIIB
III
III
1q21+
VRd
sCR
0
1.39E−03
0
0
Alive at 19mo
P4
F
47y
IgG-λ
IIIA
II
II
Normal
VRd
VGPR
0
1.13E−02
0
2.83E−05
Alive at 18mo
P5
F
58y
IgA-λ
IIIA
I
I
1q21+
VRd
sCR
0
0
0
0
Alive at 20mo
P6
F
64y
IgG-λ+λ
IIIA
II
II
t(4;14),1q21+
VRd
sCR
0
5.20E−04
0
0
Alive at 14mo
P7
M
39y
IgG-λ
IIIA
I
II
t(4;14),1q21+
VRd
VGPR
1.80E−04
5.32E−04
0
1.10E−04
Alive at 12mo
DS Durie–Salmon, ISS international staging system, R-ISS revised international staging system, ASCT autologous stem cell transplant, VRd bortezomib, lenalidomide and dexamethasone, sCR stringent complete response, VGPR very good partial response

4 Discussion

Therapeutic approaches for treating MM have advanced to include novel drugs, particularly immunotherapies. The combined use of proteasome inhibitors, thalidomide analogs, and CD38-targeting monoclonal antibodies currently represents the mainstay of modern myeloma therapy. New monoclonal antibodies, T-cell activators, and cell therapy are also in the process of entering the clinics. Although a definite cure for MM is still lacking, the introduction of new drugs with different mechanisms and improved treatment approaches has significantly improved the survival of MM patients [27]. MRD has a strong predictive value in various disease states and treatment conditions [28, 29]. It can identify the likelihood of relapse and enable early intervention. Evaluation of MRD rates is also used as an endpoint to accelerate drug testing and approval in many trials [30, 31].
Although many methods, such as NGF and multi-parametric flow cytometry, can be used to detect MRD, there is no standard method. At present, NGF is the most common method used for detecting MRD in clinics. MRD detection based on NGF is fast, efficient, and economical; however, it requires complex visualization and professional data analysis. Furthermore, false-negative MRD detection can occur in some patients due to immunophenotypic changes post-treatment [32].
The use of NGS for detecting MRD has increasingly been implemented in clinical practice. A study has compared NGS and NGF in detecting MRD and concluded a strong correlation between the two approaches [33].
MM patients in China tend to be younger than those in Europe or the United States. Furthermore, there is a high demand for effective management of MM in this population. However, the detection of MRD in Chinese MM patients has primarily relied on flow cytometry, which is gradually becoming insufficient to meet patient needs. Although international studies have previously reported on MRD monitoring via second-generation sequencing, there is limited research on the Chinese population.
To the best of our knowledge, this study is the first to compare NGS with NGF in China. To monitor MRD by using NGS, the clonal IGH rearrangement at the time of diagnosis must be known. In this study, both NGS and CE were used to detect the clonal IGH rearrangements in 60 newly diagnosed Chinese MM patients, and the consistency between the two methods was 98.3%. The overall detection rate of the IGH-FR1/FR2/FR3 combination was 70.0% via NGS. Additionally, unique clonal IGH rearrangements were observed in 42 patients. Therefore, NGS could detect clonal rearrangements in most of the newly diagnosed MM patients. Such detection can serve as a molecular biomarker at the time of diagnosis, enabling MRD monitoring during clinical treatment. To evaluate the feasibility of NGS in follow-up MRD monitoring of MM patients, we analyzed the limit and repeatability of NGS in detecting MRD. According to the IMWG guidelines, MM patients are considered MRD-negative if there are no clonal plasma cells in the bone marrow, with a minimum sensitivity of 1 in 105 nucleated cells via the NGS method [14]. The results of the study presented here confirmed that NGS has good sensitivity in MRD detection, and demonstrated a linear curve ranging from 10–6 to 10–1, with a correlation coefficient of 0.985. Using this method, it is possible to detect one tumor plasma cell in 1,000,000 nucleated cells, indicating a limit of detection of 10–6. Thus, NGS exhibits high sensitivity in MRD detection in MM patients. In addition, this approach showed good repeatability in MRD detection in these patients. In samples with different tumor loads, the MRD levels were estimated at 10–2, 10–3, and 10–4 via NGS, and the intra- and inter-assay variation was relatively low.
Currently, the major approaches recommended for MRD assessment in MM patients at home and abroad are the multi-parameter NGF and NGS technologies. There are relatively many reports on multi-parameter NGF in MRD detection in MM patients [34, 35], whereas the applicability of NGS has seldom been reported in China yet. In this study, 43 samples from 36 patients were evaluated at follow-up by using both NGS and NGF. Our results revealed a consistency rate of 79.1% between the two methods, showing that both methods have high consistency. Interestingly, out of the cases analyzed, 9 showed inconsistent MRD results, with MRD levels being detectable via NGS but undetectable via NGF. It is worth noting that none of the samples identified as MRD-positive via NGF were found to be negative via NGS. Discrepancies between NGF and NGS in detecting MRD can be attributed to differences in sensitivity and detection principles. NGF, which relies on antibodies targeting cell surface proteins, and NGS, which identifies genetic mutations, focus on distinct biological markers [36, 37]. This divergence in methodological focus can lead to scenarios where MRD is detectable by one technique but remains undetected by the other, reflecting the distinct detection capabilities inherent to each method [38].
MM patients undergoing CAR-T therapy targeting MM surface antigens, such as CD138 and CD229, may experience blocking of these antigen-binding sites for several months [39], This necessitates adjustments in the use of NGF for MRD detection. Interestingly, after ASTC, 2 patients tested MRD-negative via NGF. However, the NGS method revealed MRD levels of 2.83 × 10–5 and 1.10 × 10–4 in these patients. Notably, both patients demonstrated a very good partial response according to the evaluation of treatment effectiveness following treatment with VRd. Previous studies have also reported [40] that after induction treatment or transplantation, MRD that turns negative indicates a better clinical prognosis.
Retrospective studies have shown that making treatment decisions based on MRD results (including stopping, intensifying, or changing the treatment) can improve progression-free survival in comparison with patients whose treatment remains unmodified after MRD assessment [31, 32, 41]. The prognostic value of MRD, as determined by NGS, offers a robust basis for informed treatment adjustments, encompassing de-escalation, intensification, or modification strategies to halt disease progression and improve outcomes [29, 41]. Furthermore, by uncovering the genetic and immunologic drivers of MRD, NGS facilitates the development of targeted therapies, advancing personalized medicine in MM. This transformative approach not only promises improved therapeutic efficacy and patient well-being but also significantly shifts the MM management paradigm [41]. Although NGS presents a higher per-sample costs, its superior sensitivity in detecting MRD at very low levels offers potential long-term cost savings. Early and accurate MRD detection can guide more effective treatment adjustments, potentially reducing the overall treatment costs by avoiding unnecessary therapies and hospitalizations.
Emerging technologies, especially the integration of artificial intelligence (AI) and machine learning (ML) are set to enhance MRD detection in MM by processing complex datasets more efficiently, automating the identification of novel MRD markers, and enabling personalized treatment plans through predictive modeling [42, 43]. Concurrently, the discovery of new biomarkers such as extracellular matrix proteins, angiogenic factors, p53-related protein kinase, circulating tumor cells, and microRNAs is redefining MM diagnosis and treatment [4448]. The future of MM management is geared towards integrating these technologies and biomarkers into a personalized, predictive, and patient-centered care framework.

5 Limitation

The present study has several major limitations, including its retrospective design, the small number of cases and the intrinsic differences between two methods. The sample size was largely dependent on the number of suitable samples available from the biological sample bank, which had been collected from patients treated in a previous study within a specific timeframe. Additionally, the follow-up period was relatively short. In future studies, we intend to address these limitations by expanding the sample size, extending the follow-up duration, and conducting analyses of overall or progression-free survival. It is crucial to conduct prospective studies to confirm the feasibility and utility of NGS in MRD monitoring. Acknowledging the limitations of NGS and NGF technologies is crucial in advancing MRD detection in MM. Issues such as sample quality, with DNA integrity vital for NGS and cell viability for NGF, can impact sensitivity and accuracy, while potential contamination during processing may lead to false positives, affecting clinical decisions [36, 49]. Strict lab protocols help mitigate contamination risks. NGS’s susceptibility to errors in complex genomic regions and NGF’s potential to miss MRD due to antigenic shifts post-treatment highlight inherent technology limitations.

6 Conclusion

NGS can be used to detect clonality of IGH rearrangements in the majority of Chinese patients newly diagnosed with MM. Clonality of IGH rearrangements can be used as a molecular biomarker at the time of diagnosis for MRD monitoring after clinical treatment. NGS can be used to detect MRD with high specificity, sensitivity, and repeatability in Chinese MM patients during the follow-up period. However, the correlation between these experimental results and clinical outcomes remains to be confirmed with more samples in clinical practice.

Declarations

The study was conducted in accordance with the Declaration of Helsinki and was approved by the ethics committee of the First Affiliated Hospital of Soochow University. Informed consent was obtained from all individual participants included in the study. All the samples were from Multiple myeloma Biobank of Hematologic Biobank, National Clinical Research Center for Hematologic Diseases, Jiangsu Biobank of Clinical Resources.
Informed consent was obtained from all individual participants included in the study.

Competing interests

The authors declare no conflict of interest.
Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://​creativecommons.​org/​licenses/​by/​4.​0/​.

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Unsere Produktempfehlungen

e.Med Interdisziplinär

Kombi-Abonnement

Für Ihren Erfolg in Klinik und Praxis - Die beste Hilfe in Ihrem Arbeitsalltag

Mit e.Med Interdisziplinär erhalten Sie Zugang zu allen CME-Fortbildungen und Fachzeitschriften auf SpringerMedizin.de.

e.Med Innere Medizin

Kombi-Abonnement

Mit e.Med Innere Medizin erhalten Sie Zugang zu CME-Fortbildungen des Fachgebietes Innere Medizin, den Premium-Inhalten der internistischen Fachzeitschriften, inklusive einer gedruckten internistischen Zeitschrift Ihrer Wahl.

Anhänge

Supplementary Information

Below is the link to the electronic supplementary material.
Literatur
1.
Zurück zum Zitat Rajkumar SV, Dimopoulos MA, Palumbo A, et al. International myeloma working group updated criteria for the diagnosis of multiple myeloma. Lancet Oncol. 2014;15(12):e538-548.PubMedCrossRef Rajkumar SV, Dimopoulos MA, Palumbo A, et al. International myeloma working group updated criteria for the diagnosis of multiple myeloma. Lancet Oncol. 2014;15(12):e538-548.PubMedCrossRef
2.
Zurück zum Zitat Disease GBD, Injury I, Prevalence C. Global, regional, and national incidence, prevalence, and years lived with disability for 328 diseases and injuries for 195 countries, 1990–2016: a systematic analysis for the global burden of disease study 2016. Lancet. 2017;390(10100):1211–59.CrossRef Disease GBD, Injury I, Prevalence C. Global, regional, and national incidence, prevalence, and years lived with disability for 328 diseases and injuries for 195 countries, 1990–2016: a systematic analysis for the global burden of disease study 2016. Lancet. 2017;390(10100):1211–59.CrossRef
3.
4.
5.
Zurück zum Zitat Wang S, Xu L, Feng J, et al. Prevalence and Incidence of multiple myeloma in urban area in China: a national population-based analysis. Front Oncol. 2019;9:1513.PubMedCrossRef Wang S, Xu L, Feng J, et al. Prevalence and Incidence of multiple myeloma in urban area in China: a national population-based analysis. Front Oncol. 2019;9:1513.PubMedCrossRef
6.
Zurück zum Zitat Lu J, Lu J, Chen W, et al. Clinical features and treatment outcome in newly diagnosed Chinese patients with multiple myeloma: results of a multicenter analysis. Blood Cancer J. 2014;4(8): e239.PubMedPubMedCentralCrossRef Lu J, Lu J, Chen W, et al. Clinical features and treatment outcome in newly diagnosed Chinese patients with multiple myeloma: results of a multicenter analysis. Blood Cancer J. 2014;4(8): e239.PubMedPubMedCentralCrossRef
7.
Zurück zum Zitat Facon T, Dimopoulos MA, Meuleman N, et al. A simplified frailty scale predicts outcomes in transplant-ineligible patients with newly diagnosed multiple myeloma treated in the FIRST (MM-020) trial. Leukemia. 2020;34(1):224–33.PubMedCrossRef Facon T, Dimopoulos MA, Meuleman N, et al. A simplified frailty scale predicts outcomes in transplant-ineligible patients with newly diagnosed multiple myeloma treated in the FIRST (MM-020) trial. Leukemia. 2020;34(1):224–33.PubMedCrossRef
8.
Zurück zum Zitat Olszewski AJ, Dusetzina SB, Eaton CB, Davidoff AJ, Trivedi AN. Subsidies for oral chemotherapy and use of immunomodulatory drugs among medicare beneficiaries with myeloma. J Clin Oncol. 2017;35(29):3306–14.PubMedPubMedCentralCrossRef Olszewski AJ, Dusetzina SB, Eaton CB, Davidoff AJ, Trivedi AN. Subsidies for oral chemotherapy and use of immunomodulatory drugs among medicare beneficiaries with myeloma. J Clin Oncol. 2017;35(29):3306–14.PubMedPubMedCentralCrossRef
9.
Zurück zum Zitat Leleu X, Gorsh B, Bessou A, et al. Survival outcomes for patients with multiple myeloma in France: a retrospective cohort study using the Systeme National des Donnees de Sante national healthcare database. Eur J Haematol. 2023;111(1):125–34.PubMedCrossRef Leleu X, Gorsh B, Bessou A, et al. Survival outcomes for patients with multiple myeloma in France: a retrospective cohort study using the Systeme National des Donnees de Sante national healthcare database. Eur J Haematol. 2023;111(1):125–34.PubMedCrossRef
10.
Zurück zum Zitat Swan D, Lynch K, Gurney M, O’Dwyer M. Current and emerging immunotherapeutic approaches to the treatment of multiple myeloma. Ther Adv Hematol. 2019;10:2040620719854171.PubMedPubMedCentralCrossRef Swan D, Lynch K, Gurney M, O’Dwyer M. Current and emerging immunotherapeutic approaches to the treatment of multiple myeloma. Ther Adv Hematol. 2019;10:2040620719854171.PubMedPubMedCentralCrossRef
11.
Zurück zum Zitat Martinez-Lopez J, Lahuerta JJ, Pepin F, et al. Prognostic value of deep sequencing method for minimal residual disease detection in multiple myeloma. Blood. 2014;123(20):3073–9.PubMedPubMedCentralCrossRef Martinez-Lopez J, Lahuerta JJ, Pepin F, et al. Prognostic value of deep sequencing method for minimal residual disease detection in multiple myeloma. Blood. 2014;123(20):3073–9.PubMedPubMedCentralCrossRef
12.
Zurück zum Zitat van Dongen JJ, Langerak AW, Bruggemann M, et al. Design and standardization of PCR primers and protocols for detection of clonal immunoglobulin and T-cell receptor gene recombinations in suspect lymphoproliferations: report of the BIOMED-2 concerted action BMH4-CT98-3936. Leukemia. 2003;17(12):2257–317.PubMedCrossRef van Dongen JJ, Langerak AW, Bruggemann M, et al. Design and standardization of PCR primers and protocols for detection of clonal immunoglobulin and T-cell receptor gene recombinations in suspect lymphoproliferations: report of the BIOMED-2 concerted action BMH4-CT98-3936. Leukemia. 2003;17(12):2257–317.PubMedCrossRef
13.
Zurück zum Zitat Faham M, Zheng J, Moorhead M, et al. Deep-sequencing approach for minimal residual disease detection in acute lymphoblastic leukemia. Blood. 2012;120(26):5173–80.PubMedPubMedCentralCrossRef Faham M, Zheng J, Moorhead M, et al. Deep-sequencing approach for minimal residual disease detection in acute lymphoblastic leukemia. Blood. 2012;120(26):5173–80.PubMedPubMedCentralCrossRef
14.
Zurück zum Zitat 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):e328–46.PubMedCrossRef 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):e328–46.PubMedCrossRef
15.
Zurück zum Zitat Short NJ, Jabbour E, Albitar M, et al. Recommendations for the assessment and management of measurable residual disease in adults with acute lymphoblastic leukemia: a consensus of North American experts. Am J Hematol. 2019;94(2):257–65.PubMedCrossRef Short NJ, Jabbour E, Albitar M, et al. Recommendations for the assessment and management of measurable residual disease in adults with acute lymphoblastic leukemia: a consensus of North American experts. Am J Hematol. 2019;94(2):257–65.PubMedCrossRef
16.
Zurück zum Zitat Arcila ME, Yu W, Syed M, et al. 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. 2019;21(2):330–42.PubMedPubMedCentralCrossRef Arcila ME, Yu W, Syed M, et al. 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. 2019;21(2):330–42.PubMedPubMedCentralCrossRef
17.
Zurück zum Zitat Yao WQ, Zhu MQ, Yan LZ, et al. Clinical implication of minimal residual disease monitoring by 10-color flow cytometry in multiple myeloma. Chin J Hematol. 2019;40(9):720–5. Yao WQ, Zhu MQ, Yan LZ, et al. Clinical implication of minimal residual disease monitoring by 10-color flow cytometry in multiple myeloma. Chin J Hematol. 2019;40(9):720–5.
18.
Zurück zum Zitat van Krieken JH, Langerak AW, Macintyre EA, et al. Improved reliability of lymphoma diagnostics via PCR-based clonality testing: report of the BIOMED-2 concerted action BHM4-CT98-3936. Leukemia. 2007;21(2):201–6.PubMedCrossRef van Krieken JH, Langerak AW, Macintyre EA, et al. Improved reliability of lymphoma diagnostics via PCR-based clonality testing: report of the BIOMED-2 concerted action BHM4-CT98-3936. Leukemia. 2007;21(2):201–6.PubMedCrossRef
19.
Zurück zum Zitat Langerak AW, Groenen PJ, Bruggemann M, et al. EuroClonality/BIOMED-2 guidelines for interpretation and reporting of Ig/TCR clonality testing in suspected lymphoproliferations. Leukemia. 2012;26(10):2159–71.PubMedPubMedCentralCrossRef Langerak AW, Groenen PJ, Bruggemann M, et al. EuroClonality/BIOMED-2 guidelines for interpretation and reporting of Ig/TCR clonality testing in suspected lymphoproliferations. Leukemia. 2012;26(10):2159–71.PubMedPubMedCentralCrossRef
20.
Zurück zum Zitat Tembhare PR, Yuan CM, Venzon D, et al. Flow cytometric differentiation of abnormal and normal plasma cells in the bone marrow in patients with multiple myeloma and its precursor diseases. Leuk Res. 2014;38(3):371–6.PubMedCrossRef Tembhare PR, Yuan CM, Venzon D, et al. Flow cytometric differentiation of abnormal and normal plasma cells in the bone marrow in patients with multiple myeloma and its precursor diseases. Leuk Res. 2014;38(3):371–6.PubMedCrossRef
21.
Zurück zum Zitat Arroz M, Came N, Lin P, et al. Consensus guidelines on plasma cell myeloma minimal residual disease analysis and reporting. Cytometry B Clin Cytom. 2016;90(1):31–9.PubMedCrossRef Arroz M, Came N, Lin P, et al. Consensus guidelines on plasma cell myeloma minimal residual disease analysis and reporting. Cytometry B Clin Cytom. 2016;90(1):31–9.PubMedCrossRef
22.
Zurück zum Zitat Roshal M, Flores-Montero JA, Gao Q, et al. MRD detection in multiple myeloma: comparison between MSKCC 10-color single-tube and EuroFlow 8-color 2-tube methods. Blood Adv. 2017;1(12):728–32.PubMedPubMedCentralCrossRef Roshal M, Flores-Montero JA, Gao Q, et al. MRD detection in multiple myeloma: comparison between MSKCC 10-color single-tube and EuroFlow 8-color 2-tube methods. Blood Adv. 2017;1(12):728–32.PubMedPubMedCentralCrossRef
23.
Zurück zum Zitat Gargis AS, Kalman L, Bick DP, et al. Good laboratory practice for clinical next-generation sequencing informatics pipelines. Nat Biotechnol. 2015;33(7):689–93.PubMedPubMedCentralCrossRef Gargis AS, Kalman L, Bick DP, et al. Good laboratory practice for clinical next-generation sequencing informatics pipelines. Nat Biotechnol. 2015;33(7):689–93.PubMedPubMedCentralCrossRef
24.
Zurück zum Zitat Yao Q, Bai Y, Orfao A, Chim CS. Standardized minimal residual disease detection by next-generation sequencing in multiple myeloma. Front Oncol. 2019;9:449.PubMedPubMedCentralCrossRef Yao Q, Bai Y, Orfao A, Chim CS. Standardized minimal residual disease detection by next-generation sequencing in multiple myeloma. Front Oncol. 2019;9:449.PubMedPubMedCentralCrossRef
25.
Zurück zum Zitat Chng WJ, Dispenzieri A, Chim CS, et al. IMWG consensus on risk stratification in multiple myeloma. Leukemia. 2014;28(2):269–77.PubMedCrossRef Chng WJ, Dispenzieri A, Chim CS, et al. IMWG consensus on risk stratification in multiple myeloma. Leukemia. 2014;28(2):269–77.PubMedCrossRef
26.
Zurück zum Zitat Yao L, Chen Y, Zhai YY, et al. Characteristics of immunoglobulin heavy-chain gene clonal rearrangements by next-generation sequencing of patients with multiple myeloma. Chin J Hematol. 2021;42(8):683–6. Yao L, Chen Y, Zhai YY, et al. Characteristics of immunoglobulin heavy-chain gene clonal rearrangements by next-generation sequencing of patients with multiple myeloma. Chin J Hematol. 2021;42(8):683–6.
28.
Zurück zum Zitat Bertamini L, D’Agostino M, Gay F. MRD assessment in multiple myeloma: progress and challenges. Curr Hematol Malig Rep. 2021;16(2):162–71.PubMedCrossRef Bertamini L, D’Agostino M, Gay F. MRD assessment in multiple myeloma: progress and challenges. Curr Hematol Malig Rep. 2021;16(2):162–71.PubMedCrossRef
29.
Zurück zum Zitat 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):5988–99.PubMedPubMedCentralCrossRef 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):5988–99.PubMedPubMedCentralCrossRef
31.
Zurück zum Zitat Burgos L, Puig N, Cedena MT, et al. Measurable residual disease in multiple myeloma: ready for clinical practice? J Hematol Oncol. 2020;13(1):82.PubMedPubMedCentralCrossRef Burgos L, Puig N, Cedena MT, et al. Measurable residual disease in multiple myeloma: ready for clinical practice? J Hematol Oncol. 2020;13(1):82.PubMedPubMedCentralCrossRef
32.
Zurück zum Zitat Davies FE. Is molecular remission the goal of multiple myeloma therapy? Hematol Am Soc Hematol Educ Program. 2017;2017(1):205–11.CrossRef Davies FE. Is molecular remission the goal of multiple myeloma therapy? Hematol Am Soc Hematol Educ Program. 2017;2017(1):205–11.CrossRef
33.
Zurück zum Zitat Medina A, Puig N, Flores-Montero J, et al. Comparison of next-generation sequencing (NGS) and next-generation flow (NGF) for minimal residual disease (MRD) assessment in multiple myeloma. Blood Cancer J. 2020;10(10):108.PubMedPubMedCentralCrossRef Medina A, Puig N, Flores-Montero J, et al. Comparison of next-generation sequencing (NGS) and next-generation flow (NGF) for minimal residual disease (MRD) assessment in multiple myeloma. Blood Cancer J. 2020;10(10):108.PubMedPubMedCentralCrossRef
34.
Zurück zum Zitat 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):1159–60.PubMedCrossRef 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):1159–60.PubMedCrossRef
35.
Zurück zum Zitat Lahuerta JJ, Paiva B, Vidriales MB, et al. Depth of response in multiple myeloma: a pooled analysis of three PETHEMA/GEM clinical trials. J Clin Oncol. 2017;35(25):2900–10.PubMedCrossRef Lahuerta JJ, Paiva B, Vidriales MB, et al. Depth of response in multiple myeloma: a pooled analysis of three PETHEMA/GEM clinical trials. J Clin Oncol. 2017;35(25):2900–10.PubMedCrossRef
36.
Zurück zum Zitat Flores-Montero J, Sanoja-Flores L, Paiva B, et al. Next generation flow for highly sensitive and standardized detection of minimal residual disease in multiple myeloma. Leukemia. 2017;31(10):2094–103.PubMedPubMedCentralCrossRef Flores-Montero J, Sanoja-Flores L, Paiva B, et al. Next generation flow for highly sensitive and standardized detection of minimal residual disease in multiple myeloma. Leukemia. 2017;31(10):2094–103.PubMedPubMedCentralCrossRef
37.
Zurück zum Zitat Ha J, Lee H, Shin S, et al. 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. 2022;24(1):48–56.PubMedCrossRef Ha J, Lee H, Shin S, et al. 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. 2022;24(1):48–56.PubMedCrossRef
38.
Zurück zum Zitat Paiva B, San-Miguel J, Avet-Loiseau H. MRD in multiple myeloma: does CR really matter? Blood. 2022;140(23):2423–8.PubMedCrossRef Paiva B, San-Miguel J, Avet-Loiseau H. MRD in multiple myeloma: does CR really matter? Blood. 2022;140(23):2423–8.PubMedCrossRef
39.
Zurück zum Zitat Radhakrishnan SV, Luetkens T, Scherer SD, et al. CD229 CAR T cells eliminate multiple myeloma and tumor propagating cells without fratricide. Nat Commun. 2020;11(1):798.ADSPubMedPubMedCentralCrossRef Radhakrishnan SV, Luetkens T, Scherer SD, et al. CD229 CAR T cells eliminate multiple myeloma and tumor propagating cells without fratricide. Nat Commun. 2020;11(1):798.ADSPubMedPubMedCentralCrossRef
40.
Zurück zum Zitat 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;132(23):2456–64.PubMedPubMedCentralCrossRef 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;132(23):2456–64.PubMedPubMedCentralCrossRef
41.
Zurück zum Zitat Martinez-Lopez J, Alonso R, Wong SW, et al. Making clinical decisions based on measurable residual disease improves the outcome in multiple myeloma. J Hematol Oncol. 2021;14(1):126.PubMedPubMedCentralCrossRef Martinez-Lopez J, Alonso R, Wong SW, et al. Making clinical decisions based on measurable residual disease improves the outcome in multiple myeloma. J Hematol Oncol. 2021;14(1):126.PubMedPubMedCentralCrossRef
42.
Zurück zum Zitat Chabane K, Charlot C, Gugenheim D, et al. Real life evaluation of AlphaMissense predictions in hematological malignancies. Leukemia. 2024;38(2):420–3.PubMedCrossRef Chabane K, Charlot C, Gugenheim D, et al. Real life evaluation of AlphaMissense predictions in hematological malignancies. Leukemia. 2024;38(2):420–3.PubMedCrossRef
43.
Zurück zum Zitat Zeng Q, Klein C, Caruso S, et al. Artificial intelligence-based pathology as a biomarker of sensitivity to atezolizumab–bevacizumab in patients with hepatocellular carcinoma: a multicentre retrospective study. Lancet Oncol. 2023;24(12):1411–22.PubMedCrossRef Zeng Q, Klein C, Caruso S, et al. Artificial intelligence-based pathology as a biomarker of sensitivity to atezolizumab–bevacizumab in patients with hepatocellular carcinoma: a multicentre retrospective study. Lancet Oncol. 2023;24(12):1411–22.PubMedCrossRef
44.
Zurück zum Zitat Gupta N, Sharma A, Sharma A. Emerging biomarkers in multiple myeloma: a review. Clin Chim Acta. 2020;503:45–53.PubMedCrossRef Gupta N, Sharma A, Sharma A. Emerging biomarkers in multiple myeloma: a review. Clin Chim Acta. 2020;503:45–53.PubMedCrossRef
45.
Zurück zum Zitat Pula A, Robak P, Robak T. MicroRNA in multiple myeloma—a role in pathogenesis and prognostic significance. Curr Med Chem. 2021;28(33):6753–72.PubMedCrossRef Pula A, Robak P, Robak T. MicroRNA in multiple myeloma—a role in pathogenesis and prognostic significance. Curr Med Chem. 2021;28(33):6753–72.PubMedCrossRef
46.
Zurück zum Zitat Hideshima T, Cottini F, Nozawa Y, et al. p53-related protein kinase confers poor prognosis and represents a novel therapeutic target in multiple myeloma. Blood. 2017;129(10):1308–19.PubMedPubMedCentralCrossRef Hideshima T, Cottini F, Nozawa Y, et al. p53-related protein kinase confers poor prognosis and represents a novel therapeutic target in multiple myeloma. Blood. 2017;129(10):1308–19.PubMedPubMedCentralCrossRef
47.
Zurück zum Zitat van de Donk N. How we manage newly diagnosed multiple myeloma with circulating tumor cells. J Clin Oncol. 2023;41(7):1342–9.PubMedCrossRef van de Donk N. How we manage newly diagnosed multiple myeloma with circulating tumor cells. J Clin Oncol. 2023;41(7):1342–9.PubMedCrossRef
48.
Zurück zum Zitat Dutta AK, Alberge JB, Lightbody ED, et al. MinimuMM-seq: genome sequencing of circulating tumor cells for minimally invasive molecular characterization of multiple myeloma pathology. Cancer Discov. 2023;13(2):348–63.PubMedCrossRef Dutta AK, Alberge JB, Lightbody ED, et al. MinimuMM-seq: genome sequencing of circulating tumor cells for minimally invasive molecular characterization of multiple myeloma pathology. Cancer Discov. 2023;13(2):348–63.PubMedCrossRef
49.
Zurück zum Zitat Piskorz AM, Ennis D, Macintyre G, et al. Methanol-based fixation is superior to buffered formalin for next-generation sequencing of DNA from clinical cancer samples. Ann Oncol. 2016;27(3):532–9.PubMedCrossRef Piskorz AM, Ennis D, Macintyre G, et al. Methanol-based fixation is superior to buffered formalin for next-generation sequencing of DNA from clinical cancer samples. Ann Oncol. 2016;27(3):532–9.PubMedCrossRef
Metadaten
Titel
Evaluation of next-generation sequencing versus next-generation flow cytometry for minimal-residual-disease detection in Chinese patients with multiple myeloma
verfasst von
Mo Zhou
Yan Chen
Yanlei Gong
Mingqing Zhu
Jiannong Cen
Jinlan Pan
Lingzhi Yan
Jingjing Shang
Song Jin
Xiaolan Shi
Weiqin Yao
Shuang Yan
Depei Wu
Suning Chen
Chengcheng Fu
Li Yao
Publikationsdatum
01.12.2024
Verlag
Springer US
Erschienen in
Discover Oncology / Ausgabe 1/2024
Print ISSN: 1868-8497
Elektronische ISSN: 2730-6011
DOI
https://doi.org/10.1007/s12672-024-00938-w

Weitere Artikel der Ausgabe 1/2024

Discover Oncology 1/2024 Zur Ausgabe

Alphablocker schützt vor Miktionsproblemen nach der Biopsie

16.05.2024 alpha-1-Rezeptorantagonisten Nachrichten

Nach einer Prostatabiopsie treten häufig Probleme beim Wasserlassen auf. Ob sich das durch den periinterventionellen Einsatz von Alphablockern verhindern lässt, haben australische Mediziner im Zuge einer Metaanalyse untersucht.

Mammakarzinom: Senken Statine das krebsbedingte Sterberisiko?

15.05.2024 Mammakarzinom Nachrichten

Frauen mit lokalem oder metastasiertem Brustkrebs, die Statine einnehmen, haben eine niedrigere krebsspezifische Mortalität als Patientinnen, die dies nicht tun, legen neue Daten aus den USA nahe.

Labor, CT-Anthropometrie zeigen Risiko für Pankreaskrebs

13.05.2024 Pankreaskarzinom Nachrichten

Gerade bei aggressiven Malignomen wie dem duktalen Adenokarzinom des Pankreas könnte Früherkennung die Therapiechancen verbessern. Noch jedoch klafft hier eine Lücke. Ein Studienteam hat einen Weg gesucht, sie zu schließen.

Viel pflanzliche Nahrung, seltener Prostata-Ca.-Progression

12.05.2024 Prostatakarzinom Nachrichten

Ein hoher Anteil pflanzlicher Nahrung trägt möglicherweise dazu bei, das Progressionsrisiko von Männern mit Prostatakarzinomen zu senken. In einer US-Studie war das Risiko bei ausgeprägter pflanzlicher Ernährung in etwa halbiert.

Update Onkologie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.