How to quantify monoclonal free light chains in plasma cell disorders: which mass spectrometry technology?
Editorial Commentary

How to quantify monoclonal free light chains in plasma cell disorders: which mass spectrometry technology?

Caroline Moreau1,2, Charles R. Lefevre1,2, Olivier Decaux3,4

1Biochemistry Laboratory, Pontchaillou Hospital CHU Rennes, Rennes, France;2Univ Rennes, CHU Rennes, INSERM, EHESP, IRSET (Institut de Recherche en Santé, Environment et Travail) UMR_S 1085, Rennes, France;3Internal Medicine, CHU Rennes, Rennes, France;4Hematology, Pontchaillou Hospital CHU Rennes, Rennes, France

Correspondence to: Caroline Moreau. Laboratoire de Biochimie-Toxicologie, Hôpital Pontchaillou – CHU Rennes, 2, rue Henri Le Guilloux, 35033 Rennes, France. Email: Caroline.MOREAU@chu-rennes.fr.

Provenance and Peer Review: This article was commissioned and reviewed by the Guest Section Editor Dr. Ying Zhao (Department of Laboratory Medicine, the First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China).

Comment on: Sepiashvili L, Kohlhagen MC, Snyder MR, et al. Direct Detection of Monoclonal Free Light Chains in Serum by Use of Immunoenrichment-Coupled MALDI-TOF Mass Spectrometry. Clin Chem 2019;65:1015-22.


Submitted Mar 02, 2020. Accepted for publication Mar 13, 2020.

doi: 10.21037/atm.2020.03.200


Plasma cell disorders are a cytogenetically heterogeneous group of hematologic diseases characterized by altered clonal plasma cell proliferation, the presence of a monoclonal protein (M-protein) in serum or urine, and/or the presence of monoclonal plasma cells in the bone marrow. Multiple myeloma (MM) is the malignant stage of these disorders, which is usually preceded by monoclonal gammopathy of undetermined significance (MGUS) which is an asymptomatic premalignant stage of MM. This stage has been noted in approximately 4% of the population over 50 years of age (1-3). The rate of progression from MGUS to MM is 0.5% to 1% per year. Smoldering MM (SMM) is an intermediary stage between MGUS and MM, with a risk of progression to MM of approximately 10% per year in the first 5 years after diagnosis (2).

The diagnosis of plasma cell disorders is based on clinical, imaging, and laboratory criteria. For laboratory testing of serum, the detection and quantification of a M-protein are essential to make diagnosis. Serum protein electrophoresis (PEL) is traditionally performed and allows relative quantification of the M-protein by integration of the area under the curve of the peak representing the monoclonal entity. Serum immunofixation electrophoresis (IFE) can then be used to determine the M-protein isotype. Finally, serum quantification of free light chains (sFLC) is necessary to diagnose and monitor patients with plasma cell disorders (1). The recommendations of the International Myeloma Working Group (IMWG) and most of the published studies have been based on sFLC measured using the first commercialized assay developed in 2001, namely the Freelite® assay (The Binding Site Group Ltd, UK) (4). This assay allows sFLC quantification using a nephelometric or turbidimetric analyzer, and it is based on sheep polyclonal anti-kappa and anti-lambda FLC antibodies. However, other commercialized assays have become available. The N LatexTM sFLC kappa and lambda assay (Siemens Healthineers Diagnostics GmbH, Germany) (5) is an immunonephelometry assay based on noncompeting mouse monoclonal antibodies. The SeraliteTM assay (Sébia, Evry, France) (6) is a lateral flow immunoassay based on competitive inhibition. The Sébia Free Light Chain AssayTM (Sébia, Evry, France) is a sandwich enzyme-linked immunosorbent assay using rabbit polyclonal antibodies against free kappa or lambda light chains in the capture phase, and rabbit polyclonal horseradish peroxidase-conjugated antibodies directed against kappa or lambda light chains (7). The Diazyme Human kappa and lambda FLC assayTM (Diazyme Laboratories Inc., Poway CA, USA) is based on rabbit polyclonal antibodies, which is a latex particle-enhanced immunoturbidimetric assay. However, none of these assays directly quantifies monoclonal sFLC. Instead, the clonality is indirectly assessed by the quantification of kappa and lambda sFLC and the interpretation of the sFLC kappa/lambda ratio. Indeed, the use of a kappa/lambda sFLC ratio appears to be a more sensitive marker of monoclonal FLC production than the absolute sFLC concentration, because the interpretation incorporates the suppression of the non-tumor sFLC. Moreover, the nephelometric/turbidimetric assays present numerous technical limitations: lot-to-lot variations, antigen excess, nonlinear responses especially at higher dilution, poor post-dilution linearity, detection and quantification of oligomerization of monoclonal sFLC, gap in quantification, under and overestimation with extreme values, a huge heterogeneity in FLC measurements illustrated in pairwise comparison with external quality controls, different results on the same samples according to the analytical platform used, and the reference ranges for the Freelite® test being dependent on the analytical platform (8,9). Regarding the Sebia FLC assay, technical experience reveals that several analytical improvements such as dilutions need to be made (10). No standard reference material is available to compare assays and analytical platforms, which is a major limitation of sFLC assays. And that is the reason why new commercialized assays are compared with the Freelite® assay, which is the first commercialized assay and the one used in establishment of guidelines (5,10-12). Numerous discrepancies between tests have been documented for patients especially with chronic kidney disease and AL amyloidosis. For example, while an extended kappa/lambda sFLC ratio has been proposed for patients with chronic kidney disease when the sFLC are evaluated with the Freelite® assay (13), this is not a requirement with the N Latex assay (8,14,15). But with a worsening of renal function, the Sebia Free Light Chain Assay detected an increase in both serum kappa and lambda FLC, and an increase in the kappa/lambda sFLC ratio (16). A number of studies have concluded that the various sFLC assays are safe but not commutable, as the numerical results can vary drastically for individual patients, while the qualitative results and the trends in monitoring are in agreement (8,10-12,17). Furthermore, all recommendations were established with Freelite® assay. So, in AL amyloidosis patients or when predicting of risk of progression from SMM to MM, the IMWG guidelines specify that the values refer to the Freelite® assay (1). Consequently, the other tests cannot be used for staging, especially in clinical trials, irrespective of the performance of these tests (11,18,19). Nevertheless, few studies to date have documented the performance of other sFLC assays in these conditions (10,17).

The use of mass spectrometry to detect M-proteins in serum and urine is a recent development, and its aim is to provide a greater degree of analytical sensitivity and specificity. Since 2014, scientists have sought to develop mass spectrometry for use in the diagnosis and management of plasma cell disorders and to demonstrate that mass spectrometry can advance the way that plasma cell disorders are screened, diagnosed, and monitored. Most of the publications to date have been based on MALDI-TOF methods with immunoenrichment (Table 1), and since the initial development of mass spectrometry development for use with plasma cell disorders, MALDI-TOF MS has consistently exhibited superior sensitivity and specificity compared to conventional methods. In mass spectrometry, the main advantage is that the compound of interest, in this case monoclonal immunoglobulins or monoclonal sFLC, is defined by both the retention time and the m/z ratio. Contrary to commercialized sFLC assays, mass spectrometry allows direct quantification of the monoclonal protein, without excess of antigen or nonlinearity issues, with high sensitivity to detect monoclonal protein over the polyclonal background (Table 1). However, previous studies have demonstrated abnormal FLC ratios in samples in which the FLC were measured with MALDI-TOF mass spectrometry, thus suggesting that the methods still need to be improved.

Table 1
Table 1 Published mass spectrometry methods for the detection and quantification of monoclonal proteins in plasma cell disorders
Full table

Indeed, in a recent study published in Clinical Chemistry titled “Direct Detection of Monoclonal Free Light Chains in serum by use of immunoenrichment-coupled MALDI-TOF Mass Spectrometry”, Sepiashvili et al. sought to develop an analytically sensitive method for direct detection of monoclonal sFLC that was independent of the sFLC kappa/lambda ratio, thereby allowing conventional biochemistry techniques to be bypassed (29). The development of mass spectrometry to quantify sFLC has taken place in a series of steps. Initially, micro-LC-electrospray ionization-quadrupole-TOF-MS was shown to exhibit greater analytical sensitivity and specificity than PEL and IFE techniques (22,25). Methods were then developed for higher throughput approaches for routine clinical analyses that involved total immunoglobulin immunoenrichment with reduction and total light chain mass measurement (MASS-SCREEN and MASS-FIX) by MALDI-TOF. The analytical sensitivity of these methods was comparable to IFE. However, in patient’s plasma samples, there was a high abundance of light chains from intact immunoglobulins, and the challenge was to distinguish monoclonal sFLC against the background from light chains associated with intact monoclonal immunoglobulin and/or polyclonal immunoglobulins. Moreover, an abnormal sFLC ratio in MASS-SCREEN-negative samples was noted in previous studies (20), suggesting that specific considerations are required for sFLC measurements. To address this issue, the authors developed sFLC enrichment coupled with MALDI-TOF MS methods (FLC MALDI-TOF MS). One hundred sixty-seven sera were selected based on the presence of M-protein as determined by IFE and the sFLC ratio and these were then used to quantify sFLC with FLC MALDI-TOF MS. The results were compared with IFE, MASS-FIX, and the sFLC ratio determined with Freelite® FLC.

With specific immunoenrichment, the overall agreement of FLC MALDI-TOF MS with the sFLC ratio was 98% for sera with a low (lambda-positive) kappa/lambda sFLC ratio and for sera with normal (negative) kappa/lambda sFLC ratio. For sera with high kappa/lambda sFLC ratio (kappa-positive), the overall agreement was 79%. One hundred percent of cases with abnormal sFLC ratios with detectable M-protein on IFE were confirmed with FLC MALDI-TOF MS and 76% of cases with abnormal sFLC ratios undetectable by IFE. FLC MALDI-TOF MS allows better identification of sera with lower sFLC ratios, which enhanced the confirmation of the monoclonal sFLC by 43% compared with IFE. However, 24% of the M-proteins were identified by the sFLC ratio rather than by FLC MALDI-TOF MS. The authors suggest the increased of the binding capacity or the analytical sensitivity and resolution for optimization of the preanalytical processing. Furthermore, the authors demonstrated that FLC MALDI-TOF MS can detect monoclonal sFLC at lower concentrations of monoclonal sFLC than IFE and other mass spectrometry techniques. FLC immunoenrichment eliminated a significant proportion of the polyclonal background than could have masked the presence of monoclonal sFLC.

Finally, mass spectrometry is presently the only methodology that allows detection and quantification of monoclonal sFLC independently of the κ/λ sFLC ratio. Although the FLC immunoenrichment method eliminates the polyclonal background, the sensitivity of the method needs to be increased in order to identify all monoclonal sFLC. Preanalytical optimization is still needed to improve the performance of sFLC extraction.

FCL immunoenrichment coupled to MALDI-TOF MS is an attractive approach, although a number of considerations should be taken into account. These comprise its feasibility in routine laboratories, optimization of the manual preanalytical process, cost, and technical and clinical limitations. The clinical performance of the method should now be evaluated in patients with monoclonal gammopathy with or without renal impairment, in AL amyloidosis patients, while the pertinence of the test to assess treatment response, disease recurrence, and residual disease should also be evaluated. And one question remains unanswered: what is the place of this technique in clinical trials whose IMWG recommendations are based on the use of the Freelite assay?

Despite the quantitative results of sFLC, the authors state that the methodology is essentially based on qualitative detection of monoclonal sFLC and that quantification approaches should be developed for monoclonal sFLC.

Finally, the method published by Sepiashvili et al. appears to be the culmination of years of study (Table 1) that ultimately resulted in the development of what is now generally considered to be the best mass spectrometry method to detect sFLC in plasma cell disorders.


Acknowledgments

Funding: None.


Footnote

Conflicts of Interest: All authors have completed the ICMJE uniform disclosure form (available at http://dx.doi.org/10.21037/atm.2020.03.200). OD reports grants from Sébia, grants from Siemens, grants from Binding site, outside the submitted work; CM reports grants from Sebia, grants from Binding Site, grants from Siemens, outside the submitted work. These grants were used in independent clinical trials to buy assays to tests all assays commercialized to measure free light chains. CRL has no conflicts of interest to declare.

Ethical Statement: The authors are accountable for all aspects of the work in ensuring that questions related to the accuracy or integrity of any part of the work are appropriately investigated and resolved.

Open Access Statement: This is an Open Access article distributed in accordance with the Creative Commons Attribution-NonCommercial-NoDerivs 4.0 International License (CC BY-NC-ND 4.0), which permits the non-commercial replication and distribution of the article with the strict proviso that no changes or edits are made and the original work is properly cited (including links to both the formal publication through the relevant DOI and the license). See: https://creativecommons.org/licenses/by-nc-nd/4.0/.


References

  1. Rajkumar SV, Dimopoulos MA, Palumbo A, et al. International Myeloma Working Group updated criteria for the diagnosis of multiple myeloma. Lancet Oncol 2014;15:e538-48. [Crossref] [PubMed]
  2. Kyle RA, San-Miguel JF, Mateos MV, et al. Monoclonal gammopathy of undetermined significance and smoldering multiple myeloma. Hematol Oncol Clin North Am 2014;28:775-90. [Crossref] [PubMed]
  3. Kyle RA, Therneau TM, Rajkumar SV, et al. Prevalence of monoclonal gammopathy of undetermined significance. N Engl J Med 2006;354:1362-9. [Crossref] [PubMed]
  4. Bradwell AR, Carr-Smith HD, Mead GP, et al. Highly sensitive, automated immunoassay for immunoglobulin free light chains in serum and urine. Clin Chem 2001;47:673-80. [Crossref] [PubMed]
  5. te Velthuis H, Knop I, Stam P, et al. N Latex FLC - new monoclonal high-performance assays for the determination of free light chain kappa and lambda. Clin Chem Lab Med 2011;49:1323-32. [Crossref] [PubMed]
  6. Campbell JP, Heaney JLJ, Shemar M, et al. Development of a rapid and quantitative lateral flow assay for the simultaneous measurement of serum κ and λ immunoglobulin free light chains (FLC): inception of a new near-patient FLC screening tool. Clin Chem Lab Med 2017;55:424-34. [Crossref] [PubMed]
  7. Jacobs JFM, de Kat Angelino CM, Brouwers HMLM, et al. Evaluation of a new free light chain ELISA assay: bringing coherence with electrophoretic methods. Clin Chem Lab Med 2018;56:312-22. [Crossref] [PubMed]
  8. Moreau C, Autier B, Cavey T, et al. Evaluation of the Impact of Renal Failure on Correlation and Concordance Between 2 Free Light Chain Assays. Clin Lymphoma Myeloma Leuk 2016;16:693-704. [Crossref] [PubMed]
  9. Jacobs JFM, Tate JR, Merlini G. Is accuracy of serum free light chain measurement achievable? Clin Chem Lab Med 2016;54:1021-30. [Crossref] [PubMed]
  10. Caillon H, Avet-Loiseau H, Attal M, et al. Comparison of Sebia Free Light Chain Assay With Freelite Assay for the Clinical Management of Diagnosis, Response, and Relapse Assessment in Multiple Myeloma. Clin Lymphoma Myeloma Leuk 2019;19:e228-37. [Crossref] [PubMed]
  11. Hoedemakers RMJ, Pruijt JFM, Hol S, et al. Clinical comparison of new monoclonal antibody-based nephelometric assays for free light chain kappa and lambda to polyclonal antibody-based assays and immunofixation electrophoresis. Clin Chem Lab Med 2011;50:489-95. [PubMed]
  12. Carr-Smith HD, Jenner EL, Evans JAR, et al. Analytical issues of serum free light chain assays and the relative performance of polyclonal and monoclonal based reagents. Clin Chem Lab Med 2016;54:997-1003. [Crossref] [PubMed]
  13. Hutchison CA, Harding S, Hewins P, et al. Quantitative assessment of serum and urinary polyclonal free light chains in patients with chronic kidney disease. Clin J Am Soc Nephrol 2008;3:1684-90. [Crossref] [PubMed]
  14. Jacobs JFM, Hoedemakers RMJ, Teunissen E, et al. N Latex FLC serum free light-chain assays in patients with renal impairment. Clin Chem Lab Med 2014;52:853-9. [Crossref] [PubMed]
  15. Kennard A, Hawley C, Tate J, et al. Comparison of Freelite™ and N Latex serum free light chain assays in subjects with end stage kidney disease on haemodialysis. Clin Chem Lab Med 2016;54:1045-52. [Crossref] [PubMed]
  16. Lutteri L, Jacobs JFM. Reference ranges of the Sebia free light chain ratio in patients with chronic kidney disease. Clin Chem Lab Med 2018;56:e232-4. [Crossref] [PubMed]
  17. Henriot B, Rouger E, Rousseau C, et al. Prognostic value of involved/uninvolved free light chain ratio determined by Freelite and N Latex FLC assays for identification of high-risk smoldering myeloma patients. Clin Chem Lab Med 2019;57:1397-405. [Crossref] [PubMed]
  18. Lock RJ, Saleem R, Roberts EG, et al. A multicentre study comparing two methods for serum free light chain analysis. Ann Clin Biochem 2013;50:255-61. [Crossref] [PubMed]
  19. Mollee P, Tate J, Pretorius CJ. Evaluation of the N Latex free light chain assay in the diagnosis and monitoring of AL amyloidosis. Clin Chem Lab Med 2013;51:2303-10. [Crossref] [PubMed]
  20. Milani P, Murray DL, Barnidge DR, et al. The utility of MASS-FIX to detect and monitor monoclonal proteins in the clinic. Am J Hematol 2017;92:772-9. [Crossref] [PubMed]
  21. Kohlhagen MC, Barnidge DR, Mills JR, et al. Screening Method for M-Proteins in Serum Using Nanobody Enrichment Coupled to MALDI-TOF Mass Spectrometry. Clin Chem 2016;62:1345-52. [Crossref] [PubMed]
  22. Barnidge DR, Dispenzieri A, Merlini G, et al. Monitoring free light chains in serum using mass spectrometry. Clin Chem Lab Med 2016;54:1073-83. [Crossref] [PubMed]
  23. Mills JR, Kohlhagen MC, Dasari S, et al. Comprehensive Assessment of M-Proteins Using Nanobody Enrichment Coupled to MALDI-TOF Mass Spectrometry. Clin Chem 2016;62:1334-44. [Crossref] [PubMed]
  24. Mills JR, Barnidge DR, Murray DL. Detecting monoclonal immunoglobulins in human serum using mass spectrometry. Methods 2015;81:56-65. [Crossref] [PubMed]
  25. Barnidge DR, Krick TP, Griffin TJ, et al. Using matrix-assisted laser desorption/ionization time-of-flight mass spectrometry to detect monoclonal immunoglobulin light chains in serum and urine. Rapid Commun Mass Spectrom 2015;29:2057-60. [Crossref] [PubMed]
  26. VanDuijn MM, Jacobs JFM, Wevers RA, et al. Quantitative measurement of immunoglobulins and free light chains using mass spectrometry. Anal Chem 2015;87:8268-74. [Crossref] [PubMed]
  27. Botz CM, Barnidge DR, Murray DL, et al. Detecting monoclonal light chains in urine: microLC-ESI-Q-TOF mass spectrometry compared to immunofixation electrophoresis. Br J Haematol 2014;167:437-8. [Crossref] [PubMed]
  28. Barnidge DR, Dasari S, Botz CM, et al. Using mass spectrometry to monitor monoclonal immunoglobulins in patients with a monoclonal gammopathy. J Proteome Res 2014;13:1419-27. [Crossref] [PubMed]
  29. Sepiashvili L, Kohlhagen MC, Snyder MR, et al. Direct Detection of Monoclonal Free Light Chains in Serum by Use of Immunoenrichment-Coupled MALDI-TOF Mass Spectrometry. Clin Chem 2019;65:1015-22. [Crossref] [PubMed]
Cite this article as: Moreau C, Lefevre CR, Decaux O. How to quantify monoclonal free light chains in plasma cell disorders: which mass spectrometry technology? Ann Transl Med 2020;8(15):973. doi: 10.21037/atm.2020.03.200

Download Citation