Whole Exome Sequencing of Aberrant Plasma Cells in a Patient with Multiple Myeloma Minimal Residual Disease

2017 ◽  
Vol 30 (Suppl 2) ◽  
pp. 2S75-2S80
Author(s):  
Martina Zátopková ◽  
Jana Filipová ◽  
Tomáš Jelínek ◽  
Petr Vojta ◽  
Tereza Ševčíková ◽  
...  
Blood ◽  
2018 ◽  
Vol 132 (Supplement 1) ◽  
pp. 4956-4956
Author(s):  
Weiqin Yao ◽  
Zhu Mingqing ◽  
Yao Feirong ◽  
Lingzhi Yan ◽  
Song Jin ◽  
...  

Abstract Objective: In the last decade the outcome in multiple myeloma in CHINA has greatly improved due to the new, effective therapies including PIs and Imids. But responses to treatment and survival remains heterogeneous because of patient characteristic, disease biology and mechanisms of drug resistance. More and more studies have established the link between depth of response and improved PFS and OS. multiparameter-flow cytometry (MFC) is a main method to detect minimal residual disease(MRD) in myeloma. Sensitivity will be at least at 10-4 to 10-5 by 10-color MFC. Imaging techniques such as PET-CT are important for EMD and bone MRD detection. whole body DWI-MRI is a new imaging technique by mean of the apparent diffusion coefficient(ADC) which can qualify the depth of response to antineoplastic treatment. This study was designed to evaluate the prognostic significance of MRD by 10-color MFC and imaging to the MM patients after induction.Methods: 102 patients with newly diagnosed MM were enrolled at the First Affiliated Hospital of Soochow University from July 2015 to July 2017. All patients were diagnosed and the response were assessed by IMWG criteria. The median of age was 58 (31-75).There were 46 patients with IgG type , 24 IgA , 14 light chain, 18 others. 34 Patients in ISS stageⅠ,34 in stage Ⅱ, 30 in stage Ⅲ. All patients received 4-6 cycles of triplet bortezomib based or lenalidomide based induction therapy. Transplantation available patients received APBSCT with BUCY condition followed by 4-6 cycles of bortezomib based or lenalidomide based consolidation which were given to transplantation unavailable patients too. Lenalidomide and thalidomide were used for over 2y of maintenance therapy. Bone marrow aspirates for MRD imaging MRD assessment were obtained at the end of induction and 1year after ASCT.The median of follow-up was 13 (2-29) months.Results: According to MRD by MFC and imaging after induction therapy and 1 year after ASCT, the patients were divided into different groups. MFC negativity was 33%(29/88) after induction therapy compared with 63%(32/51) after ASCT (X2=11.636,P=0.001). After induction therapy, the median PFS was 22 months for MRD positive group compared with not reached with MRD negative group by MFC (P=0.042) in patients with very good partial remission(VGPR) and above. The 2 years PFS was 100% for those with MRD negative compared with 60% for MRD positive by imaging. The 2 years PFS was 80% for those have multiclonal normal plasma cells compared with 52.6% for those without. The median PFS was not reached for MFC MRD negative patients 1 year after ASCT compared with 20 months for positive patients. (P=0.002). Multivariate analysis including high risk cytogenetics(17p-, t(4;14), t(14;16)), sex, age, ISS, chemotherapy, ASCT, CR/VGPR, normal PCs showed that the MFC MRD and ASCT were independent prognostic factor.Conclusions: Patients with MFC MRD negative after induction therapy or ASCT is a better prognostic marker than CR or even the best marker. Imaging MRD negativity and the appearance of normal plasma cells in the bone marrow suggests a better prognosis.We will have a try to do more research on overall survival(OS),include longer follow-up and a larger number of patients enrolled. Figure. Figure. Disclosures No relevant conflicts of interest to declare.


2020 ◽  
Vol 9 (11) ◽  
pp. 3519
Author(s):  
Elena Zamagni ◽  
Paola Tacchetti ◽  
Simona Barbato ◽  
Michele Cavo

The International Myeloma Working Group (IMWG) recently introduced the evaluation of minimal residual disease (MRD) within the multiple myeloma (MM) response criteria, and MRD negativity assessed inside and outside the bone marrow is currently considered the most powerful predictor of favorable long-term outcomes. However, MRD evaluation has thus far relied on flow-cytometry or molecular-based methods, despite the limitations associated with the patchy infiltration of bone marrow (BM) plasma cells and the presence of extra-medullary (EMD). On the contrary, imaging-based sensitive response assessment through the use of functional rather than morphological whole-body (WB) imaging techniques, such as positron emission tomography with computed tomography (PET/CT) and magnetic resonance imaging (MRI), likely is a promising strategy to overcome these limitations in evaluating response to therapy and in the assessment of the MRD status in MM patients. However, despite the significant advances in the development and availability of novel functional imaging techniques for MRD evaluation, a worldwide standardization of imaging criteria for acquisition, interpretation, and reporting is yet to be determined and will be object of future investigations.


2021 ◽  
Vol 11 (1) ◽  
Author(s):  
Takeshi Yoroidaka ◽  
Kentaro Narita ◽  
Hiroyuki Takamatsu ◽  
Momoko Fujisawa ◽  
Shinji Nakao ◽  
...  

AbstractIn this study, the minimal residual disease (MRD) levels in patients with multiple myeloma (MM) were assessed by comparing the new 8-color single-tube multiparameter flow cytometry method (DuraClone), which reduces the cost of antibodies and labor burden of laboratories, with the EuroFlow next-generation flow (NGF) method. A total of 96 samples derived from 69 patients with MM were assessed to determine the total cell acquisition number (tCAN), percentages of total and normal plasma cells (PCs), and MRD levels using two methods. We found that the tCAN was significantly higher with EuroFlow-NGF than with DuraClone (median 8.6 × 106 vs. 5.7 × 106; p < 0.0001). In addition, a significant correlation in the MRD levels between the two methods was noted (r = 0.92, p < 0.0001). However, in the qualitative analysis, 5.2% (5/96) of the samples showed discrepancies in the MRD levels. In conclusion, the DuraClone is a good option to evaluate MRD in multiple myeloma but it should be used with caution.


Blood ◽  
2008 ◽  
Vol 112 (11) ◽  
pp. 5141-5141
Author(s):  
Lucie Kovarova ◽  
Ivana Buresova ◽  
Ludek Pour ◽  
Jaroslav Michalek ◽  
Roman Hajek

Abstract Background: Multiple myeloma (MM) is a hematological malignancy characterized by expansion of neoplastic plasma cells (PCs). Despite of advances in treatment, achieved in past ten years, many patients relapse due to minimal residual disease (MRD) existence. Aim: Monitoring of number and phenotype of plasma cells in transplanted MM patients by multiparametric flow cytometry. Patients and Methods: Bone marrow of 24 MM patients (13 male, 11 female) after autologous stem cells transplantation was used. Analyses were done in 1st month and 1st year after transplantation. Relapsed patients were analyzed earlier. Plasma cells were identified as CD138+CD38+ cells, CD19 and CD56 markers were used for discrimination of normal polyclonal (N-PCs) and abnormal monoclonal (A-PCs) plasma cells, respectively. Results: Median number of PCs in whole group of patients in 1st month after transplantation was 0.1% (range 0.0–2.5%), median number of CD19+ N-PC 39.6% (5.0–79.2%), CD56+ A-PC 31.7% (3.5–90.9%) and N-PC/A-PC ratio was 1.1 (0.1–22.6). There were identified 8 relapsed patients during 1st year from transplantation, all with detectable monoclonal immunoglobulin, and with median number of PCs 10.6% (range 0.1–76.6%), N-PC 0.2% (0.0–56.2%), A-PC 60.1% (0.8–99.9%) and N-PC/A-PC ratio 0.0 (0.0–3.0). In a group of non-relapsed patients in 1st year after transplantation was median number of PCs 0.2% (range 0.0–1.9%), N-PC 64.2% (6.3–90.0%), A-PC 21.7% (3.0–93.2%) and N-PC/A-PC ratio 3.4 (0.1–25.0). There is an evidence that relapse is relate to loss of CD19 expression on PCs and to higher number of CD56+ PC. Conclusion: Flow cytometric analysis of PCs phenotype profile is relatively quick and sufficiently sensitive method that can be used for monitoring of MRD. Detailed analysis of higher number of patients and comparison of their data with other parameters (immunofixation etc.) are necessary for verification of this method and for its application in routine diagnostics.


2020 ◽  
Vol 3 (1) ◽  
Author(s):  
Claudia Tapia-Alveal ◽  
Timothy R. Olsen ◽  
Tilla S. Worgall

AbstractMultiple myeloma (MM) is a neoplasm of plasma cells that secrete patient specific monoclonal immunoglobulins. A recognized problem in MM treatment is the early recognition of minimal residual disease (MRD), the major cause of relapse. Current MRD detection methods (multiparameter flow cytometry and next generation sequencing) are based on the analysis of bone marrow plasma cells. Both methods cannot detect extramedullary disease and are unsuitable for serial measurements. We describe the methodology to generate high affinity DNA aptamers that are specific to a patient’s monoclonal Fab region. Such aptamers are 2000-fold more sensitive than immunofixation electrophoresis and enabled detection and quantification of MRD in serum when conventional MRD methods assessed complete remission. The aptamer isolation process that requires small volumes of serum is automatable, and Fab specific aptamers are adaptable to multiple diagnostic formats including point-of-care devices.


Blood ◽  
2016 ◽  
Vol 128 (22) ◽  
pp. 237-237
Author(s):  
Karma Salem ◽  
Jihye Park ◽  
Claudia Freymond ◽  
Marzia Capelletti ◽  
Daisy Huynh ◽  
...  

Abstract Introduction: Recent data shows that multiple myeloma (MM) almost always arises from precursor states called Monoclonal Gammopathy of Undetermined Significance (MGUS) and Smoldering Multiple Myeloma (SMM), but not all patients with MGUS or SMM develop MM. Risk factors of progression for SMM patients are largely based on tumor load as represented by an M-protein ≥ 3 g/dL, a free light chain (FLC) ratio outside the range of 0.125 to 8, and ≥ 10% plasma cells in the BM. However, the genetic lesions that underlie progression, the molecular factors that cause rapid versus slow progression, and the factors that distinguish the relatively indolent MGUS from SMM are not well known. Further, the genomic landscape of SMM is not well characterized. One potential factor is MYC overexpression. Bergsagel et al. have found that MYC levels increase when comparing MGUS, SMM and overt MM. Other frequently altered pathways in MM are NF-kB, MAPK and DNA damage. In addition, limited studies of paired SMM and MM samples show that in many cases, the aggressive subclones can already be detected, in small cell fractions, before overt MM develops. However, the cause of progression to MM is unclear, in large part because sequential genomic studies of MGUS/SMM progression have yet to be undertaken. To address these questions, in this study we examine clinically-annotated samples from patients with SMM. Methods: We performed whole exome sequencing (WES) (mean target coverage 50X/100X) on 49 germline-tumor matched samples from patients with SMM (DNA from bone marrow CD138+ plasma cells matched with germline DNA from peripheral blood mononuclear cells). Libraries were constructed using Agilent SureSelect XT2 library prep kit, and hybridized to Agilent's whole exome V5+UTR capture probes and then sequenced on HiSeq 2500 (Illumina). We also performed targeted deep sequencing using a custom enrichment bait set on 25 samples of progressor (n=12) and non-progressor (n=13) SMM samples. Libraries were also constructed with Agilent SureSelect XT2 library prep kit and enriched by hybridizing to an in-house designed customized target bait, then sequenced on HiSeq 2500. Sequencing data were analyzed using previously established analytic pipelines including MuTect, RecapSeg, GISTIC, MutSig, and ABSOLUTE. Results: The number of Somatic Single Nucleotide Variants (SSNVs) seen in SMM ranged from 1 to 98 nonsilent mutations with an average of 1.14 mutations/Mb, which is slightly lower than MM (1.6 mutations/Mb) from previous studies (p-value=0.05). This large and varying range of mutational load among samples suggests that SMM is likely a heterogenous entity where some patients are closer to MGUS and others closer to MM. We identified likely drivers in SMM in about ~32% of the samples, including mutations in MM candidate driver genes such as NRAS, KRAS and PTPN11(overall 36 events were present in COSMIC). SMM also had somatic CNAs in about ~50% of SMM samples, such as hyperdiploidy, gain of chromosome 1q, deletion of 13p and 17p, which match the hallmark chromosome changes seen in MM. Comparing deep targeted sequencing of 100 genes (mean target coverage 361X) in samples from 12 SMM patients who progressed to myeloma vs. 13 SMM patients who did not, we found non-synonymous mutations exclusive to progressors, suggesting that with more samples we may find genetic alterations that predict progression in SMM. Conclusion: This study demonstrates that WES and targeted sequencing of SMM patients can detect MM candidate driver genes as well as hallmark CNAs seen in MM patients. Further, there may be potential different mutational features between progressors and non-progressors. Thus, this approach can be used to identify genetic drivers of clonal progression from MGUS/SMM to MM that may present opportunities for early therapeutic intervention and prevention of disease progression. Disclosures Roccaro: Takeda Pharmaceutical Company Limited: Honoraria. Ghobrial:Takeda: Honoraria; Noxxon: Honoraria; Amgen: Honoraria; Novartis: Honoraria; BMS: Honoraria, Research Funding; Celgene: Honoraria, Research Funding.


Blood ◽  
2019 ◽  
Vol 134 (Supplement_1) ◽  
pp. 1799-1799
Author(s):  
Marie C Bene ◽  
Nelly Robillard ◽  
Philippe Moreau ◽  
Soraya Wuilleme

Background. During the follow-up of treated myeloma patients, the assessment of minimal residual disease (MRD) is gaining an increasing importance. The detection of remaining abnormal plasma-cells (PC) may rely on molecular techniques investigating immunoglobulin rearrangements of the malignant clone or on multiparameter flow cytometry (MFC). The latter allows to obtain a rapid response by dealing with fresh cells. It also focuses on cells still alive, since dead cells are discarded as debris. Numerous publications have reported that the most reliable markers of PC in MFC are CD38 and CD138 their co-expression being a good way to select the population of PC in a bone marrow (BM) or, more rarely tested, blood sample. Malignant PC often but not always differ from normal PC by the loss of CD19 expression and the acquisition of CD56. Other immunophenotypic alterations are related, among others, to the expression of CD20, CD27, CD28, CD33, CD45, CD81 or CD117. Malignant PC also display the monotypic usage of light chains by the myelomatous immunoglobulin, which can readily be assessed in MFC after permeabilization of the PC, although this induces an additional technical step that could induce some cell loss. Here we compared the two panels proposed by the Euroflow consortium (Flores Montero, 2018) which use the same backbone of antibodies with a "surface" strategy associating CD81 and CD117 or a "cytoplasmic" strategy investigating for the expression of kappa and lambda immunoglobulin light chains. Methods. From a cohort of patients for whom MRD had been assessed in our MFC platform, 100 samples were retrospectively selected as displaying detectable MRD in the cytoplasmic strategy. All BM samples had first been submitted to bulk lysis to increase the PC concentration. Between 5 and 10x106 nucleated cells were used for surface staining, premeabilization and intracytoplasmic staining. Another aliquot of the same suspension, with 3 to 5x106 nucleated cells, was used for the "surface" tube. Briefly, both samples were surface stained with antibodies to CD45 (Ozyme), CD19 (Beckman Coulter), CD38 (Cytognos), CD138 (BD Biosciences) and CD27 (Ozyme). The "surface tube" also contained antibodies to CD81 (Clinisciences) and CD117 (BD Biosciences). After this incubation, the "cytoplasmic tube" was submitted to permeabilization (Intrastain® Dako) and cells further incubated with antibodies to kappa and lambda chains (Dako and Clinisciences). All samples were acquired on the same day. Listmodes of the "cytoplasmic tubes" were analyzed and data provided to the clinician within 24 hours. For this study, the listmodes of the "surface tube" were analyzed blindly using the Kaluza® software. Data were then compared to those of the "cytoplasmic tube" Results. A good linear correlation was observed between the two results, with a R2 coefficient of 0.73. The global difference between both tubes was usually a lower MRD level detected with the "cytoplasmic tube", seen in 68% of the cases (median -0.0113; range -0.0001 to -1.4). Of note higher levels (0.0007 to 1.44) were observed in 32%, ruling out a systematic loss of cells that could have been responsible for this difference. The gating strategy adopted (Robillard 2013) delineated four populations on a CD19/CD56 bivariate histogram. Monotypy was then investigated in each of the four subsets thus identified. The same strategy was applied for the "surface tube" looking at the coexpression profile of CD81 and CD117 in each subset. Globally, 58% of the samples were CD56 positive among which 43 were CD19-. CD19 was also absent in 40 CD56- samples. All configurations of CD117 and CD81 coexpression were seen, making each patient a challenging case. In about 10% of the cases, two suspect subsets were seen in the "surface tube" while monotypy was seen in only one in the "cytoplasmic tube". Conclusion. Although this study shows a good correlation between the two panels, it was found that a greater confidence could be attributed to the "cytoplasmic tube", where data are comforted by identification of a monotypic population with the same light chain as the monoclonal peak. Moreover, although confirmation of the abnormal subset was required in numerous cases with the "surface tube", the reverse was never observed. Single use of the "cytoplasmic" combination can thus be recommended as a robust method of MRD assessment in multiple myeloma. Disclosures Moreau: AbbVie: Consultancy, Honoraria; Amgen: Consultancy, Honoraria; Janssen: Consultancy, Honoraria; Takeda: Consultancy, Honoraria; Celgene: Consultancy, Honoraria.


Sign in / Sign up

Export Citation Format

Share Document