scholarly journals Will Mass Spectrometry Replace Current Techniques for Both Routine Monitoring and MRD Detection in Multiple Myeloma?

Hemato ◽  
2021 ◽  
Vol 2 (4) ◽  
pp. 764-768
Author(s):  
Katie L. Thoren

In recent years, mass spectrometry has been increasingly used for the detection of monoclonal proteins in serum. Mass spectrometry is more analytically sensitive than serum protein electrophoresis and immunofixation, can help distinguish therapeutic monoclonal antibodies from M-proteins, and can detect the presence of post-translational modifications. Mass spectrometry also shows promise as a less-invasive, peripheral-blood-based test for detecting minimal residual disease in multiple myeloma. Studies comparing the clinical utility of mass spectrometry to current blood- and bone-marrow-based techniques have been conducted. Although still primarily limited to research settings, clinical laboratories are starting to adopt this technique for patient care. This review will discuss the current status of mass spectrometry testing for multiple myeloma, the benefits and challenges of this technique, and how it may be incorporated into clinical practice in the future.

2021 ◽  
Vol 39 (15_suppl) ◽  
pp. 8009-8009
Author(s):  
Angela Dispenzieri ◽  
Amrita Y. Krishnan ◽  
Bonnie Arendt ◽  
Surendra Dasari ◽  
Yvonne Adeduni Efebera ◽  
...  

8009 Background: Measuring response among patients with multiple myeloma is essential for the care of patients. Deeper responses have been associated with better progression free survival (PFS) and overall survival (OS). Serum (SIFE) and urine immunofixation are the currently used markers for biochemical documentation of CR after which marrow is tested for plasma cell clearance. Next generation flow cytometry and sequencing are used to document the presence of minimal residual disease (MRD). Mass spectrometry of blood by MALDI (Mass-Fix) is a new simple, inexpensive, sensitive, and specific means of detecting monoclonal immunoglobulins. To better test the hypothesis that Mass-Fix is superior to existing methodologies to predict for survival outcomes—especially SIFE-- samples from the STAMINA trial (NCT01109004), a trial comparing 3 transplant approaches among patients who have already received induction, were employed. Methods: Five-hundred and seventy-five patients were included. Samples from enrollment post-induction (post-I) and 1-year post enrollment (1YR) were tested when available. Four response parameters were assessed univariately: Mass-Fix, SIFE, complete response, and MRD by next generation flow cytometry. Mass spectrometry spectra were evaluated in a blinded fashion. Complete response was according to the 2006 International Myeloma Working Group criteria. Multivariate Cox proportional hazard models using stepwise regression were developed to explore the independent effect of the different response parameters on PFS and OS and interactions with other risk factors. Results: Of the 4 response measures, only MRD and Mass-Fix predicted for PFS and OS at multiple testing points on multivariate analyses (Table). Of the 4 post-I measurements, only MRD predicted for PFS; however, Mass-Fix was the only post-I measurement to predict for OS. Of all the 1-year measures, both 1YR Mass-Fix and 1YR MRD positivity predicted for inferior PFS and OS. In models including MRD and Mass-Fix, SIFE and CR were not prognostic for PFS or OS. Conclusions: Mass-Fix is a powerful means to track monoclonal proteins. The full utility of Mass-Fix was not exploited given the absence of a diagnostic sample and the fact that only serum (and not urine) was tested. Despite these limitations, it performed well at pre-induction and at 1 year. Mass-Fix provides a convenient and non-invasive means of predicting for myeloma outcomes. Clinical trial information: NCT01109004. [Table: see text]


2021 ◽  
Vol 39 (15_suppl) ◽  
pp. 8010-8010
Author(s):  
Noemi Puig ◽  
Bruno Paiva ◽  
Teresa Contreras ◽  
M. Teresa Cedena ◽  
Laura Rosiñol ◽  
...  

8010 Background: Analysis of minimal residual disease (MRD) in the bone marrow (BM) of patients with multiple myeloma (MM) is accepted by the IMWG to evaluate treatment efficacy and is a well-established prognostic factor. However, there is an unmet need to explore the clinical value of MRD in peripheral blood (PB). Methods: Newly diagnosed MM patients enrolled in the GEM2012MENOS65 trial received six induction (Ind) cycles of bortezomib, lenalidomide, and dexamethasone (VRD) followed by autologous stem cell transplantation (ASCT) and 2 further cycles of consolidation (Cons) with VRD. MRD was analyzed in BM using Next Generation Flow (NGF) and in serum by Mass Spectrometry (MS) using IgG/A/M, κ, λ, free κ and free λ specific beads, both after Ind, at day 100 after ASCT, and after Cons. Sequential samples from the first 184 patients were analyzed. Results: Results of both methods were in agreement (NGF+/MS+ and NGF-/MS-) in 83% of cases post-Ind (152/184), 80% post-ASCT (139/174) and 76% post-Cons (128/169). Stratifying by the log range of MRD by NGF, discordances (NGF+/MS- and NGF-/MS+) seemed to increase at the lower MRD ranges, being 22%, 21% and 19% from ≥10−5 to <10−4 and 21%, 21%, 23% at ≥x10−6(post-Ind, ASCT and Cons, respectively). Analysis of discordances showed that they could be partly explained by the higher percentages of cases found to be positive by MS as compared by NGF at part of the time-points analyzed and at each log range of MRD. From ≥10−5 to <10−4, MRD was detected by NGF in 36%, 28%, 20% of cases post-Ind, ASCT and Cons, respectively vs MS in 37%, 29%, 21% of them; at ≥x10−6, NGF was positive in 11%, 14%, 19% of cases vs MS in 23%, 19% and 16% of them. Considering NGF as a reference, the negative predictive value (NPV) of MS per MRD range (≥10−5 to <10−4 and ≥x10−6, respectively) was: post-Ind: 83% (p<0,0001), 94% (p=0,034); post-ASCT 86% (p<0,0001), 90% (p=0,022); post-Cons 89% (p<0,0001), 85% (p=0,0469). Despite these discordances, the prognostic value of each technique in terms of undetectable MRD and progression-free survival (PFS) was consistent at all time-points (Table) and further, discordant cases (NGF+/MS- and NGF-/MS+) did not display a significantly different PFS as compared to NGF-/MS- cases. Conclusions: The results of MRD assessed by NGF in BM and by MS in PB show a significant concordance and are associated with a similar prognostic value analyzed in terms of PFS. Given its high NPV, MRD in peripheral blood by MS provides a gateway for BM aspiration/biopsy and MRD assessment by NGF.[Table: see text]


2021 ◽  
Vol 9 (1) ◽  
Author(s):  
Hong Ding ◽  
Juan Xu ◽  
Zhimei Lin ◽  
Jingcao Huang ◽  
Fangfang Wang ◽  
...  

AbstractMultiple myeloma (MM) is a treatable plasma cell cancer with no cure. Clinical evidence shows that the status of minimal residual disease (MRD) after treatment is an independent prognostic factor of MM. MRD indicates the depth of post-therapeutic remission. In this review article, we outlined the major clinical trials that have determined the prognostic value of MRD in MM. We also reviewed different methods that were used for MM MRD assessment. Most important, we reviewed our current understanding of MM MRD biology. MRD studies strongly indicate that MRD is not a uniform declination of whole MM tumor population. Rather, MM MRD exhibits unique signatures of cytogenetic aberration and gene expression profiles, unlike those of MM cells before therapy. Diagnostic high-risk MM and low-risk MM exhibited a diversity of MRD features. Clonal evaluation may occur at the MRD stage in MM. The dynamics from the diagnostic MM to MRD correlate with the disease prognosis. Lastly, on the aspect of omics, we performed data-based analysis to address the biological features underlying the course of diagnostic-to-MRD MM. To summarize, the MRD stage of disease represents a critical step in MM pathogenesis and progression. Demonstration of MM MRD biology should help us to deal with the curative difficulties.


Blood ◽  
2020 ◽  
Vol 136 (Supplement 1) ◽  
pp. 40-41
Author(s):  
Celine M. Vachon ◽  
Josiah Murray ◽  
Cristine Allmer ◽  
Dirk Larson ◽  
Aaron D. Norman ◽  
...  

Introduction Monoclonal gammopathy of undetermined significant (MGUS) is a premalignant plasma cell disorder that is common in individuals over age 50. MGUS prevalence estimates to date have generally been based on results of serum electrophoresis and immunofixation (for heavy chain [HC] MGUS) and additionally, free light chain (for light chain [LC] MGUS). Mass-spectrometry assays, however, can detect lower levels of monoclonal (M) proteins, identify the isotype and provide accurate quantification of the M-protein. Given the evidence that MGUS is likely present at least 10 years prior to its detection, mass spectrometry may provide more accurate estimates of underlying MGUS. We examined the prevalence of HC-MGUS using matrix-assisted laser desorption/ionization-time of flight (MALDI-TOF) mass spectrometry among three risk groups: European Americans (EA), African Americans (AA) and first-degree relatives (FDR) of patients with multiple myeloma (MM), MGUS, chronic lymphocytic leukemia (CLL) or non-Hodgkin's lymphoma (NHL). Methods We sampled all AA and a random sample of EA participants ages 50 and older enrolled in the Mayo Clinic Biobank. We also selected unaffected first-degree relatives (limiting to ages 50 and older) from our ongoing family study of hematological malignancies. We screened serum samples by the MALDI-TOF mass spectrometry assay, which is clinically used at Mayo Clinic (known as MASS-FIX) for detection and isotypes of M-proteins. The assay uses isotype-specific nanobody enrichment coupled to MALDI-TOF mass spectrometry. For each of the risk groups, HC- MGUS prevalence was estimated by age (50-59, 60-69, 70+). Age- and sex-adjusted prevalence rates were calculated by direct standardization to the 2010 US population. Ninety-five percent confidence intervals (95% CI) for the prevalence rates and comparisons across the three groups were based on the Poisson distribution. Because free LC information was unavailable, LC-MGUS was not estimated. Results A total 327 AA, 854 EA and 792 FDR were screened for M-proteins using the MASS-FIX assay. All three groups had similar sex (42-44% male) and age distributions (mean[SD] age was 64.5[9.5] yrs., 64.4[8.8] yrs. and 64.9[10.5] yrs. for AA, EA and FDR, respectively). As previously noted using conventional detection methods, the overall prevalence of HC-MGUS was higher in the AA population (16.5% [95%CI: 12.2%, 20.8%]) and FDR (21.5% [95%CI: 18.3%, 24.7%]) than in EA (11.2% [95%CI: 8.8%, 13.5%]), both p-values &lt;0.0001. This pattern was generally seen across age groups (Table 1), with increasing prevalence with age. FDR of MM or MGUS cases had a higher prevalence (22.2% [95%CI: 18.4%, 26.0%]) than FDR of CLL or NHL cases (18.8% [95%CI: 13.3%, 24.4%]), although the difference was not statistically significant (p-value=0.82). Overall prevalence estimates of HC-MGUS using MASS-FIX were at least three-fold higher than estimates using conventional methods alone (i.e. age- and sex-adjusted prevalence in Olmsted County, MN, was 3.1% [95% CI: 2.9%-3.4%]). Conclusion We provided some of the first data on prevalence of HC-MGUS across three risk groups, using a sensitive method for detecting monoclonal proteins. The MASS-FIX assay resulted in substantially higher absolute rates of HC-MGUS (at least 3-fold higher) relative to conventional methods. Further, the prevalence of HC-MGUS among the AA and FDR was significantly higher than the EA population. Thus, the more sensitive assay identified greater absolute numbers of individuals with MGUS, but resulted in similar relationships as seen in prior studies across these risk groups. Our results have implications for future studies of the etiology of MGUS and its progression. Disclosures Dispenzieri: Janssen: Research Funding; Pfizer: Research Funding; Alnylam: Research Funding; Intellia: Research Funding; Celgene: Research Funding; Takeda: Research Funding. Kumar:Amgen: Consultancy, Other: Research funding for clinical trials to the institution, Consulting/Advisory Board participation with no personal payments, Research Funding; Janssen Oncology: Other: Research funding for clinical trials to the institution, Consulting/Advisory Board participation with no personal payments; Takeda: Other: Research funding for clinical trials to the institution, Consulting/Advisory Board participation with no personal payments; AbbVie: Other: Research funding for clinical trials to the institution, Consulting/Advisory Board participation with no personal payments; Karyopharm: Consultancy; Genecentrix: Consultancy; Cellectar: Other; Carsgen: Other, Research Funding; Merck: Consultancy, Research Funding; Dr. Reddy's Laboratories: Honoraria; Adaptive Biotechnologies: Consultancy; Celgene/BMS: Other: Research funding for clinical trials to the institution, Consulting/Advisory Board participation with no personal payments; Genentech/Roche: Other: Research funding for clinical trials to the institution, Consulting/Advisory Board participation with no personal payments; Oncopeptides: Consultancy, Other: Independent Review Committee; IRC member; Kite Pharma: Consultancy, Research Funding; BMS: Consultancy, Research Funding; Tenebio: Other, Research Funding; Sanofi: Research Funding; MedImmune: Research Funding; Novartis: Research Funding. Murray:The Binding Site: Patents & Royalties: Patent Use of Mass Spec to identify monoclonal proteins licensed to The Binding Site.


2020 ◽  
Vol 66 (3) ◽  
pp. 421-433 ◽  
Author(s):  
M Zajec ◽  
P Langerhorst ◽  
M M VanDuijn ◽  
J Gloerich ◽  
H Russcher ◽  
...  

Abstract Background Monoclonal gammopathies (MGs) are plasma cell disorders defined by the clonal expansion of plasma cells, resulting in the characteristic excretion of a monoclonal immunoglobulin (M-protein). M-protein detection and quantification are integral parts of the diagnosis and monitoring of MGs. Novel treatment modalities impose new challenges on the traditional electrophoretic and immunochemical methods that are routinely used for M-protein diagnostics, such as interferences from therapeutic monoclonal antibodies and the need for increased analytical sensitivity to measure minimal residual disease. Content Mass spectrometry (MS) is ideally suited to accurate mass measurements or targeted measurement of unique clonotypic peptide fragments. Based on these features, MS-based methods allow for the analytically sensitive measurement of the patient-specific M-protein. Summary This review provides a comprehensive overview of the MS methods that have been developed recently to detect, characterize, and quantify M-proteins. The advantages and disadvantages of using these techniques in clinical practice and the impact they will have on the management of patients with MGs are discussed.


Plant Methods ◽  
2019 ◽  
Vol 15 (1) ◽  
Author(s):  
Yahui Liu ◽  
Song Lu ◽  
Kefu Liu ◽  
Sheng Wang ◽  
Luqi Huang ◽  
...  

AbstractIn recent years, mass spectrometry-based proteomics has provided scientists with the tremendous capability to study plants more precisely than previously possible. Currently, proteomics has been transformed from an isolated field into a comprehensive tool for biological research that can be used to explain biological functions. Several studies have successfully used the power of proteomics as a discovery tool to uncover plant resistance mechanisms. There is growing evidence that indicates that the spatial proteome and post-translational modifications (PTMs) of proteins directly participate in the plant immune response. Therefore, understanding the subcellular localization and PTMs of proteins is crucial for a comprehensive understanding of plant responses to biotic stress. In this review, we discuss current approaches to plant proteomics that use mass spectrometry, with particular emphasis on the application of spatial proteomics and PTMs. The purpose of this paper is to investigate the current status of the field, discuss recent research challenges, and encourage the application of proteomics techniques to further research.


Blood ◽  
2014 ◽  
Vol 124 (21) ◽  
pp. 3419-3419
Author(s):  
David L Murray ◽  
John R Mills ◽  
Jerry A. Katzmann ◽  
Robert Kyle ◽  
Vincent Rajkumar ◽  
...  

Abstract Detection of an M-protein (monoclonal immunoglobulin) has been used to diagnose and monitor multiple myeloma (MM). As therapies for MM have improved, more sensitive methods have been used to define response: immunofixation electrophoresis (IFX) of serum and urine, normalization of the serum immunoglobulin free light chain (FLC) ratio, and high sensitivity flow cytometry to detect clonal plasma bone marrow cells. It is hoped that these more sensitive approaches will differentiate those patients with minimal residual disease (MRD) versus no residual disease (NRD), that later which could mean a cure. Flow cytometry of plasma cells requires bone marrow aspiration, which is inconvenient and expensive and is potentially limited by sampling bias. More sensitive methods to differentiate MRD from NRD using serum would be advantageous. We have developed a sensitive test for the presence of the monoclonal antibody produced by the plasma cells which may serve as a substitute for invasive bone marrow biopsy. Briefly, patient serum is enriched for immunoglobulins (Ig) and the Ig light chains are decoupled from the heavy chain by reduction with DTT. The mass distribution of the light chains is resolved using a micro LC-ESI-Q-TOF mass spectrometry and the presence of the M-protein is detected as a spike in the mass distribution. In addition to the detection, the accurate mass measurement of the light chain serves as a unique individualized marker which can aid in detection in subsequent patient monitoring. We have termed this method: monoclonal-immunoglobulin-Rapid-Accurate-Mass-Measurement (miRAMM). By spiking monoclonal immunoglobulins into human serum, we have demonstrated that miRAMM is approximately 1000x more sensitive than SPEP at detecting M-proteins. Initial results on 21 patients in stringent complete response (sCR) demonstrated that 67% (n=14) had detectable malignant specific clones by miRAMM. Given these promising results we have extended the method to a larger series of myeloma patients for whom we had long term follow-up and serum samples prior to treatment and at 6-12 months status post stem cell treatment. The results of this data will be correlated to the clinical outcomes for these patients. Early results demonstrate the potential of miRAMM to be a more sensitive, cost effective approach to detect MRD compared with current methods. Disclosures Murray: Mayo Clinic: Patent Application filed Patents & Royalties. Barnidge:Mayo Clinic: Patent filed Patents & Royalties.


2016 ◽  
Vol 62 (10) ◽  
pp. 1334-1344 ◽  
Author(s):  
John R Mills ◽  
Mindy C Kohlhagen ◽  
Surendra Dasari ◽  
Patrick M Vanderboom ◽  
Robert A Kyle ◽  
...  

Abstract BACKGROUND Electrophoretic separation of serum and urine proteins has played a central role in diagnosing and monitoring plasma cell disorders. Despite limitations in resolution and analytical sensitivity, plus the necessity for adjunct methods, protein gel electrophoresis and immunofixation electrophoresis (IFE) remain front-line tests. METHODS We developed a MALDI mass spectrometry–based assay that was simple to perform, automatable, analytically sensitive, and applicable to analyzing the wide variety of monoclonal proteins (M-proteins) encountered clinically. This assay, called MASS-FIX, used the unique molecular mass signatures of the different Ig isotypes in combination with nanobody immunoenrichment to generate information-rich mass spectra from which M-proteins could be identified, isotyped, and quantified. The performance of MASS-FIX was compared to current gel-based electrophoresis assays. RESULTS MASS-FIX detected all M-proteins that were detectable by urine or serum protein electrophoresis. In serial dilution studies, MASS-FIX was more analytically sensitive than IFE. For patient samples, MASS-FIX provided the same primary isotype information for 98% of serum M-proteins (n = 152) and 95% of urine M-proteins (n = 55). MASS-FIX accurately quantified M-protein to &lt;1 g/dL, with reduced bias as compared to protein electrophoresis. Intraassay and interassay CVs were &lt;20% across all samples having M-protein concentrations &gt;0.045 g/dL, with the ability to detect M-proteins &lt;0.01 g/dL. In addition, MASS-FIX could simultaneously measure κ:λ light chain ratios for IgG, IgA, and IgM. Retrospective serial monitoring of patients with myeloma posttreatment demonstrated that MASS-FIX provided equivalent quantitative information to either protein electrophoresis or the Hevylite™ assay. CONCLUSIONS MASS-FIX can advance how plasma cell disorders are screened, diagnosed, and monitored.


2019 ◽  
Vol 19 (10) ◽  
pp. e143-e144
Author(s):  
Oscar Berlanga ◽  
Simon North ◽  
David Barnidge ◽  
Sophie Brusseau ◽  
Roshani Patel ◽  
...  

Sign in / Sign up

Export Citation Format

Share Document