Abstract

Background

Laboratory methods for diagnosis and monitoring of monoclonal gammopathies have evolved to include serum and urine protein electrophoresis, immunofixation electrophoresis, capillary zone electrophoresis, and immunosubtraction, serum-free light chain assay, mass spectrometry, and newly described QUIET.

Content

This review presents a critical appraisal of the test methods and reporting practices for the findings generated by the tests for monoclonal gammopathies. Recommendations for desirable practices to optimize test selection and provide value-added reports are presented. The shortcomings of the serum-free light chain assay are highlighted, and new assays for measuring monoclonal serum free light chains are addressed.

Summary

The various assays for screening, diagnosis, and monitoring of monoclonal gammopathies should be used in an algorithmic approach to avoid unnecessary testing. Reporting of the test results should be tailored to the clinical context of each individual patient to add value. Caution is urged in the interpretation of results of serum-free light chain assay, kappa/lambda ratio, and myeloma defining conditions. The distortions in serum-free light chain assay and development of oligoclonal bands in patients‘ status post hematopoietic stem cell transplants is emphasized and the need to note the location of original monoclonal Ig is stressed. The need for developing criteria that consider the differences in the biology of kappa and lambda light chain associated lesions is stressed. A new method of measuring monoclonal serum-free light chains is introduced. Reference is also made to a newly defined entity of light chain predominant intact immunoglobulin monoclonal gammopathy. The utility of urine testing in the diagnosis and monitoring of light chain only lesions is emphasized.

Impact statement

Multiple myeloma is the most common hematologic malignancy in adults. Autologous stem cell transplantation is used for treatment, despite controversy about improvement in overall survival.

Precursor lesions of multiple myeloma warrant monitoring despite lack of evidence for benefit of early treatment.

Serumfree light chain assay has added limited value due to lack of specificity for monoclonal light chains. MALDI-TOF MS and QUIET methods promise to improve the diagnosis and monitoring of light chain myelomas and detection of minimal residual disease. The newly described entity of light chain predominant multiple myeloma may be relevant to the consideration of shorter survival in this group and light chain escape.

Introduction and Background

Arne Wilhelm Kaurin Tiselius invented protein electrophoresis for which he was awarded a Nobel Prize in 1948. Modifications to the method include introduction of paper and agarose gel as the matrix for electrophoresis, immuno-electrophoresis, and zonal electrophoresis with immunofixation. The same methods for serum are applicable to urine protein electrophoresis, except that urine should be concentrated prior to electrophoresis (1, 2).

Diagnostic methods for monoclonal gammopathy are serum protein electrophoresis (SPEP) and serum protein immunofixation electrophoresis (SIFE). The corresponding methods for urine are urine protein electrophoresis (UPEP) and urine protein immunofixation electrophoresis (UIFE). Additional testing includes total serum and urine proteins, estimation of concentration of major protein bands, and monoclonal immunoglobulin in SPEP, by densitometry (1–9,). Capillary zone electrophoresis (CZE) and immunosubstraction provide similar information (10, 11).

There is lack of standardization about terminology for monoclonal gammopathy interpretation. The terms, paraprotein, monoclonal immunoglobulin, monoclonal band, monoclonal peak, M-protein, M-band, and M-spike are often used interchangeably. It would be preferable to use the term monoclonal immunoglobulin with immunoglobulin type (e.g., monoclonal IgG kappa). The term monoclonal free light chains is recommended for denoting the findings in light chain monoclonal gammopathy; the historical term Bence-Jones protein should be avoided (1, 8, 9).

Quantification of serum IgG, IgM, IgA, and serum-free light chains (SFLC) are in common use and recommended by the international myeloma working group (IMWG). IMWG also recommends examination of 24-hour urine. Additional tests include complete blood cell count (CBC), basic or comprehensive metabolic profile, serum levels of β2 microglobulin, and lactate dehydrogenase (LDH), for staging purposes. Bone marrow examination, cytogenetic analysis, flow cytometry and imaging studies are also useful (2, 12, 13).

Serum and urine protein electrophoreses serve as screening studies in evaluation of monoclonal gammopathy and other disorders, such as nutrition, state of hydration, acute inflammation, chronic inflammation, alpha 1 anti-trypsin deficiency, intravascular hemolysis, nephrotic syndrome, cirrhosis, iron deficiency, C3 complement, proteinuria, renal tubular damage, and cryoglobulins. The primary use of SPEP, SIFE, UPEP, and UIFE is in the diagnosis and monitoring of monoclonal gammopathies. It is recommended that the pathologist review the medical record for a comprehensive report and to promote optimal utilization of SIFE, SFLCA, and other tests. Billing codes 84165, 86334, 84166, and 86335 are used for SPEP, SIFE, UPEP, and UIFE, respectively. Including the findings from a review of the medical record is an essential first step in the effort toward reporting the results as a comprehensive clinical pathology consultation and billing under code 80502 (1–11).

There is wide variation in practice about reporting of serum and urine protein studies. At one extreme, the view is that only presence or absence of monoclonal immunoglobulins needs to be addressed, whereas pathologists opt for more comprehensive reporting with review of medical records. One argument/rationale for abbreviated reporting could be the mismatch between workload and staffing. An additional issue is the qualifications of the person signing out the protein electrophoresis reports. A pathologist has an incentive to present the SPEP and SIFE findings in the context of the clinical status of the patient, taking into account other disorders that may affect serum proteins (e.g., cirrhosis, nephrotic syndrome, acute inflammation, iron deficiency, chronic inflammatory disorders such as rheumatoid arthritis, SLE and HIV infection), and to document chart review required for a clinical consultation (14,). A PhD scientist may limit the report to presence or absence of monoclonal immunoglobulin, in part due to inability to bill for the service. Chart review may also be used by a pathologist to ensure appropriateness of testing. At one institution, the pathologist, by presenting the local utilization data, was able to convince the medical executive board that clinical staff should be limited to ordering SPEP with reflex to SIFE only, and the pathologist would decide if SIFE was warranted (14). Chart review is also relevant in distinguishing monoclonal IgG kappa bands that may result from the use of therapeutic monoclonal antibodies.

Classical monoclonal gammopathies, also referred to as neoplastic monoclonal gammopathies (NMG) consist of monoclonal gammopathy of undetermined significance (MGUS), smoldering or asymptomatic myeloma (SMM), and plasma cell myeloma/multiple myeloma (MM) (15–17).

Analysis of SPEP involves visual examination and densitometric scans, which provide the concentrations of different normal protein bands and of monoclonal immunoglobulin (MIg). Accurate quantification of MIg peaks is compromised by the lack of standard preparations of purified monoclonal Ig that could be used to calibrate the instruments. Quantitative accuracy is further complicated by the variations in the resolution of the gels used for analysis. It is recommended that a gel capable of resolving the two major bands in beta region be used as the minimal level of resolution for SPEP (1, 2, 5, 10, 11).

SIFE, UPEP, and UIFE results are evaluated by visual examination to ascertain the heavy and light chain isotype of the MIg and the presence of monoclonal free light chains. Although capillary electrophoresis (CZE) is not addressed in this review, the issues pertinent to SPEP, SIFE, UPEP, and UIFE apply to CZE as well (10).

In addition to NMGs, there are other instances in which a monoclonal immunoglobulin may be detected in serum and/or urine. MIg may be present as part of the evolution of normal immune response, with process evolving through oligoclonal pattern, one apparent MIg, polyclonal increase in immunoglobulins to normal pattern (18,). A partial list of other disorders associated with MIg includes plasmacytoma; autoimmune disorders such as chronic inflammatory demyelinating neuropathy, myasthenia gravis, cold agglutinin disease, multiple sclerosis; Waldenström macroglobulinemia, and other lymphomas; CLL and other leukemias, including plasma cell leukemia; any neoplastic disorder; AL amyloidosis; light and/or heavy chain deposition disorders; and Polyneuropathy Organomegaly Endocrinopathy Monoclonal Plasma cell disorder and Skin changes (POEMS). Therapeutic monoclonal antibodies may yield an IgG Kappa band on SPEP/SIFE (1–4, 19, 20).

The heavy chains, gamma, mu, alpha, delta, and epsilon with either kappa or lambda chains, heavy chains alone, or light chains alone could result in a monoclonal band on SPEP/SIFE. IgG kappa, IgG lambda, IgA kappa, IgA lambda intact immunoglobulins (IIg), and kappa and lambda light chain monoclonal gammopathies account for 98%–99% of the monoclonal gammopathies. The remaining monoclonal gammopathic disorders may be biclonal, rarely triclonal, nonsecretory, or with IgD or IgE specificity. For the purposes of this monograph, the term monoclonal gammopathies will be limited to Ig lesions consisting of IgG kappa, IgG lambda, IgA kappa, IgA lambda, and light chain lesions of kappa and lambda light chain specificity. Nonsecretory lesions will be addressed as appropriate. About 85% of the MM are IIg lesions, with 13%–15% being light chain lesions only, and 2%–3% represented by biclonal and nonsecretory lesions (20).

Screening and Diagnosis of NMGs

Diagnostic work-up starts with SPEP that may be a screening test or be triggered by suspicion of multiple myeloma due to lytic bone lesions, osteoporosis, pathological fracture, elevated total globulins, renal failure, unexplained neuropathy, amyloidosis, anemia, or hypercalcemia. Additional testing should be employed in an algorithmic fashion (21).

For screening, SPEP and UPEP should both be ordered (the controversy about SFLCA replacing urine testing will be addressed later). The pathologist, based on the results of SPEP and UPEP, along with review of electronic medical record (EMR), should decide if SIFE and/or UIFE and SFLCA assays are warranted. If a monoclonal band is noted on SPEP, SIFE should be done to characterize the band, to ascertain that the band is in fact MIg, and to establish its heavy and light chain specificity. Pathologic states mimicking a MIg band include fibrinogen, elevated C-reactive protein and C3c band, all in anodal gamma region. An apparent peak in the beta region due to elevated transferrin levels is usually associated with iron deficiency and sometimes triggered by estrogen treatment; an apparent peak in the alpha region due to elevated alpha 2 macroglobulin are other examples that may require SIFE to rule out MIg. An artefact at the point of application, often due to cryoglobulins, is another cause of a spurious MIg band (1–4, 14, 21). Serum immunoglobulin quantification should be done to provide information about the state of uninvolved immunoglobulins, as well as a support/corroboration for the densitometric estimate of MIg concentration.

In the case of an IIg monoclonal band in serum, UIFE should be done to ascertain the presence of monoclonal light chains in urine as the latter are nephrotoxic. UIFE should also be done if the band noted on SPEP is a light chain only band. If an IIg monoclonal band or a light chain monoclonal band are detectable by serum and urine examination, there is limited need for SFLCA, though there is wide variations in opinions (20).

In the circumstance of a monoclonal band on SPEP that does not stain for gamma, mu, or alpha heavy chains, but a light chain is detectable on SIFE, the specimen should be examined for IgD or IgE MIg. Quantification for IgD and IgE may be sufficient (1–4,). In renal failure, a monoclonal band may be detectable on SPEP when a patient has only a light chain myeloma. The likelihood of a detectable monoclonal band on SPEP is enhanced with lambda light chain myelomas due to higher tendency of lambda monoclonal light chains to dimerize (22).

Scenarios producing spurious monoclonal bands include restricted IgG subclasses (e.g., IgG4 and the previously mentioned therapeutic monoclonal antibodies) (19, 23, 24,). The low concentration of the MIg band, <0.1 g/dL, in a specific location and history of treatment with MIg is generally sufficient for identification of therapeutic MIg bands. Specific antibodies to therapeutic MIg exist in case a positive identification of a band is needed (23).

In reporting results of SPEP/SIFE, changes in non-immunoglobulin proteins should be correlated with clinical findings from review of EMR. It is not infrequent that SPEP provides the first indication of a serious disorder such as cirrhosis, nephrotic syndrome, alpha 1 anti-trypsin deficiency, chronic inflammation, and nutritional deficiency (1–7).

Reporting Findings of Electrophoretic Analyses

There is lack of standardization for reporting SPEP/SIFE, despite attempts by multiple agencies and groups. The following is a suggested format for the report when a MIg is first detected

  1. Identify the immunoglobulin type of MIg

  2. Record concentration of MIg.

  3. Record location of MIg band. Location may be recorded in general terms, such as cathodal end of gamma region, cathodal region, mid-gamma region, anodal gamma region, interface of beta-gamma region, beta region overlapping C3 band, between transferrin and C3 band, overlapping transferrin band, or alpha 2 region.

  4. The rationale for recording the location is:

    • It provides a check on the integrity of the specimen on subsequent examinations (25).

    • If an oligoclonal pattern develops following treatment, location of the neoplastic MIg band is invaluable in distinguishing between the treatment-induced bands versus recurrence or persistence of the malignant clone (26).

  5. Comment on the state of uninvolved/background immunoglobulins.

  6. Issue one consolidated report with results of SPEP, SIFE and if applicable, UPEP and UIFE. Even if a preliminary report based on SPEP alone is issued, a consolidated report with SIFE, UPEP, and UIFE results is strongly recommended.

  7. If MIg is detected for the first time and review of EMR suggests that NMG was not suspected, the pathologist should communicate directly with the attending physician so that results do not go unnoticed. This is not meant to imply that finding a monoclonal Ig constitutes a critical value, but is suggested as a value-added measure.

  8. Other clinical parameters (e.g., age, and gender of the patient, anemia, hypercalcemia, hypoalbuminemia, and major diagnoses) should be included to document review of EMR.

If MIg detected is light chain only, there is even less standardization in reporting. If the monoclonal light chain is detectable on UPEP/UIFE only, concentration of the involved SFLC may provide an estimate of the tumor mass and aid in assessing response to treatment. SFLCA does not measure the concentration of monoclonal light chains, just the mass of total light chains. Thus, using the SFLCA results for monoclonal light chain analysis is akin to using the serum levels of the involved IIg rather than the concentration of the MIg peak.

Urine specimens, whether random, first morning, or 24-h collection, should be concentrated 10 to 100-fold to enhance the sensitivity of UPEP/UIFE.

Suggested Format for Reporting if No MIg Is Detected

Even though total serum protein and concentrations of the major components (i.e., albumin, alpha 1, alpha 2, beta 1 and beta 2 and gamma globulins) are included in the routine reports, comments on abnormalities in non-Ig proteins that may be increased or decreased, genetic variants, and any prominent lipid band should be recorded. Refer to the clinical findings and make a statement supporting or contradicting a clinical differential diagnosis. Comment on SFLC results, as these may be ordered routinely, despite a high false positive and false negative rate of findings. It is imperative to caution against over-reading the abnormal kappa/lambda ratios due to their poor sensitivity and specificity for NMG disorders (20, 21, 27). Patients categorized as having MGUS solely based on an abnormal kappa/lambda ratio have been observed. It has been noted, anecdotally, that patients have been subjected to bone marrow examination to resolve an abnormal kappa/lambda ratio, without testing urine, and without any other clinical indication for additional testing.

Indications for SIFE

If no peak/band is detected on SPEP, there may still be a need to perform SIFE to rule out MIg below the detection limit of SPEP. The following are indications for SIFE:

  1. Hypogammaglobinemia: This may be a manifestation of a light chain MM.

  2. Undiagnosed polyneuropathy: Neuropathy due to an autoantibody is susceptible to treatment by plasma exchange. These monoclonal autoantibodies are usually present in too low a concentration to be discernible on SPEP.

  3. An abnormal or prominent band of normal proteins (e.g., a transferrin or C3 bands of higher than usual intensity).

  4. A broader than usual normal protein band as this may be due to an adjacent MIg band.

  5. History of lytic bone lesion, osteoporosis, or pathological fracture.

  6. History of autoimmune disorder (e.g., myasthenia gravis, cold-agglutinin disease, hemolytic anemia, or SLE).

  7. Patients status post-transplantation of hematopoietic stem cells or solid organs.

  8. History of amyloidosis.

  9. History of lymphoma or leukemia.

Appropriate rationale to avoid SIFE include lack of any of the indications listed above with normal SPEP. Polyclonal increase in gamma globulins without any suggestion of a specific peak is not an indication for SIFE, an abnormal kappa/lambda ratio notwithstanding. An abnormal kappa/lambda ratio on SFLCA per se is not an indication for SIFE (27).

If an SIFE is ordered but the attending pathologist considers it to be unwarranted, the latter may cancel the request (the negative implication for part B billing notwithstanding) and include a statement in the SPEP report that SIFE was not performed (8, 10, 14).

Indication for SIFE on Repeat Testing

Patients monitored for MGUS or SMM do not require repeat SIFE if the location of the peak has not changed. Reporting concentration of the peak without repeating SIFE is sufficient. In patients treated for MM, if MIg band is in the same location as before, reporting the concentration of MIg is sufficient. If no MIg is detectable on SPEP, SIFE is warranted as the latter has higher sensitivity. If there are one or more new bands following treatment, SIFE should be done to characterize the new bands. An oligoclonal SPEP/SIFE pattern develops in about 70% of patients treated with autologous stem cell transplantation (ASCT) (28). It may not always be feasible to distinguish between a post-treatment oligoclonal band and a recurrence/persistence of the malignant clone if both are of the same Ig type and in the same location.

In light chain MM, it is appropriate to repeat SFLCA to monitor the level of the involved light chain, keeping in mind that SFLCA does not measure monoclonal light chains only, but all light chains, benign or malignant. UIFE provides positive evidence for monoclonal light chains, if monoclonal light chain is detected (20, 27, 29).

Suggested Format for Reporting Repeat Examination by Protein Electrophoresis with Known NMG

For NMG, not being treated for multiple myeloma, a statement recording the following should suffice:

Previously reported monoclonal Ig (list type) is noted at the same location at a concentration of “x” g/dL. Concentration of the MIg was “y” g/dL on “z” date. A change in the background immunoglobulins should be documented. There is no need to perform an SIFE. However, if a urine sample was submitted a UIFE should be carried out and the results recorded in a consolidated report. It is appropriate to comment on other laboratory findings and changes in serum protein results in the context of the clinical status, as revealed by review of EMR.

For patients treated for multiple myeloma, the suggested format for repeat protein electrophoresis should include the following:

  • The previously described monoclonal Ig (mention the Ig type) is noted at a concentration of “x” g/dL. The concertation of MIg was “y” g/dL on “z” date.

  • Comment on background immunoglobulins.

  • Presence of oligoclonal bands, if any, with a description of the Ig type and location.

  • If MIg peak is noted in the same location as the original malignant spike, SIFE is not warranted. SFLCA in such patients does not add value.

  • If no MIg is detectable on SPEP, an SIFE should be performed and presence or absence of MIg documented. SFLCA may be considered to detect rare instances of light chain escape. No cases of light chain escape were observed in a 10-year, retrospective review of about 500 cases at this institution. It is postulated that the recurrence of disease in light chain predominant multiple myelomas may present with monoclonal light chains prior to the detection of monoclonal IIg thus creating the appearance of light chain escape (30).

  • In case of light chain myelomas, where no MIg was detectable on SPEP or SIFE at original examination, and no MIg is detected at repeat examination, a comment about the concentration of involved SFLC should be entered.

    • UIFE ought to be performed.

    • MALDI-TOF Mass-spectrometry or preferably quantification of monoclonal light chains by ultrafiltration immunofixation electrophoresis test (QUIET) may be performed on serum to detect and quantify monoclonal light chain (31, 32).

MIg Overlapping Normal Protein Band

If MIg clone overlaps a normal protein, usually in the beta region, it is appropriate to include a statement that MIg band overlaps C3 or transferrin and that the concentration of the 2 proteins is included in the quantitation per densitometry. Such values would be useful in monitoring the course of treatment in combination with the concentration of the involved Ig. It has been proposed that measuring heavy/light may be more accurate than trying to monitor the MIg concentration in the beta region; however, this does not seem to add meaningfully to monitoring the patient by measuring the MIg and relevant Ig concentration (33, 34).

Purported Uses of SFLCA

Screening for Monoclonal Gammopathy

The acknowledged gold standard for diagnosing monoclonal gammopathy is SPEP, SIFE, UPEP, and UIFE (35,). IMWG recommends testing for SFLCs. It has been proposed that SFLCA can obviate the need for urine testing, when employed with SPEP and SIFE. SPEP/SIFE, and using the proposed SFLC reference range of 0.26 to 1.65 does discern almost all cases of myeloma, including light chain MM. However, urine examination is also positive in 100% of the cases of light chain MM. When urine examination is positive for a monoclonal light chain, it is diagnostic of monoclonal gammopathy. An abnormal SFLCA result is not contributory in this instance and is falsely abnormal in about 36% of tertiary care patients (27,). More than 30% of patients with a detectable MIg in serum have a normal SFLCA result (20,). While using SPEP and SIFE with SFLCA will detect all significant NMGs, SFLCA will generate a large number of false positive results, entailing unneeded additional investigation, patient consternation and possible harm from unwarranted additional testing (13, 34, 36–42).

The rate of false positive kappa/lambda ratio is greater than 50% in patients with polyclonal hypergammaglobulinemia (27,). Providers sometimes diagnose patients with an abnormal κ/λ ratio, without any evidence of MIg as having MGUS and subject them to consultation by oncology (20, 27, 42, 43,). Polyclonal increase in gamma globulins may predispose to development of monoclonal gammopathy due to persistent inflammatory response (44,). Higher prevalence of kappa dominance in abnormal κ/λ ratio, without any evidence of MIg, reflects a biological preference for kappa chains. In normal serum, IgG kappa accounts for more than 60% of the total IgG. In NMGs the ratio of lesions associated with kappa vs. lambda light chains is about 2:1 (20, 27, 29, 45). This dominance of kappa preference is seen to an extreme in patients with polyclonal increase in gamma globulin. Kappa dominant, abnormal κ/λ ratio is noted in more than 90% of such cases. A similar preference for kappa chains has also been documented in the oligoclonal response noted following hematopoietic stem cell transplantation (45, 46).

The preference for kappa light chains appears to extend to NMGs. Though a normal κ/λ ratio is noted in more than 30% of the samples with a detectable MIg, this false negative rate is much higher for lambda chain associated lesions than that for kappa chain associated lesions. About 60% of kappa light chain associated MGUS samples have a normal κ/λ ratio whereas the corresponding frequency for lambda light chain associated lesions is almost 90% (20, 45,). The usefulness of SFLCA applies to monitoring of light chain myeloma and perhaps the newly defined entity of light chain predominant multiple myeloma (LCPMM). The assay could have a role in early detection of relapse in LCPMM (29, 30).

Though IMWG recommends electrophoretic evaluation of 24-h urine specimens, a concentrated random urine sample is usually adequate. First morning specimen is preferable due to it being naturally more concentrated (47).

While SFLCA is less expensive than UPEP/UIFE, SFLCA has limited utility in a screening algorithm, whereas a positive finding of MIg in UIFE is diagnostic (27, 36).

Prognostic Value of SFLC

The literature asserts that an abnormal κ/λ ratio in MGUS indicates a worse prognosis (48–53,). An abnormal κ/λ ratio is a nonspecific finding and indicates a state of chronic inflammation (53,). In patients with no MIg, an abnormal κ/λ ratio indicates a worse prognosis and the same is the case in Hodgkin disease and other lymphomas and individuals without any neoplastic disorders (51, 54,). An abnormal κ/λ ratio suggests an adverse outcome in a nonspecific way as do elevated ferritin, CRP, and RDW, and lower levels of serum albumin (52, 54–58).

Monitoring the Results of Treatment

It has been claimed that because light chains have a shorter half-life than IIg, changes in SFLCs could be used to ascertain response or lack of response to treatment faster than using level of neoplastic MIg. A retrospective review of patient records did not disclose any change in treatment based on SFLCA. A systematic examination did not reveal any benefit in using SFLCA over monitoring the level of neoplastic MIg (59, 60).

Monitoring the response of stem cell transplants with SFLCA is complicated by the emergence of oligoclonal bands in nearly 70% of cases (28, 45,). Oligoclonal bands tend to produce a kappa dominant abnormal κ/λ ratio, irrespective of the primary disorder. More than 15% of the abnormal κ/λ ratios in patients with lambda chain associated lesions (i.e., IgG lambda or IgA lambda MM), status post ASCT were kappa dominant. This is a conservative estimate of the false positive rate in that the plasma cells needed to produce sufficient excess kappa light chains to overcome the monoclonal lambda chain due to lambda chain associated malignancy, to result in kappa dominant abnormal κ/λ ratio. If this conservative 15% false positive kappa dominant abnormal κ/λ ratio is applied to patients with kappa chain associated MM, about 40% of the kappa dominant abnormal κ/λ ratios in these patients would be false positives, thus obliterating the utility of monitoring κ/λ ratio in status post ASCT patients (45, 61, 62,). There is lack of consensus about prognostic value of oligoclonal pattern (63, 64).

Following chemotherapy and ASCT, the original malignant clone may not be detectable. In such case a dominant oligoclonal band of the same immunoglobulin class may be mistaken for residual/recurrent disease. While it is helpful to note the location of the malignant Ig band to evaluate recurrence, in the absence of residual serum or electrophoretic profiles for comparison, this technique is not very accurate. The historic progression of MIg migration is made more manageable by the equipment manufacturers providing software for comparison with earlier gels (26).

It would behoove the equipment manufacturers to improve the resolution of gels to further facilitate separation of post-treatment clones from the original malignant Ig clone.

SFLCA in AL Amyloid and Non-Secretory MM

There may be a limited role for SFLCA in monitoring patients with AL amyloid and/or nonsecretory MM. Assays assessing propensity of light chains to form dimers may be a better indicator of risk for amyloidosis and in general a need for more sensitive methods has been emphasized (65–69).

Stringent Complete Response to Treatment for MM

IMWG has defined stringent complete response as meeting criteria for complete response plus a normal κ/λ ratio, however, a large number of false-positive kappa dominant abnormal κ/λ ratios with ASCT renders this criterion moot. It has been shown that lack of normal SFLCA result is not consequential to outcome of treatment (45, 70–75).

Myeloma Defining Condition

The IMWG has developed criteria for myeloma defining condition as having one of the following findings in a patient with SMM:

  • More than 60% plasma cells in bone marrow.

  • More than one bone lesion on MRI examination

  • Involved serum-free light chain concentration of greater than 100 mg/L and ratio of concentration of involved to un-involved light chain >100.

It was observed that about 16% of the SMM patients meeting criterion “c” above progressed to MM. A 16% sensitivity is poor performance for a clinical assay, and suitability of early treatment has been disputed (76–78,). The referenced study did not segregate the patients by the type of light chain. This is imperative as it has been shown that on average, kappa chain associated NMGs produce 4 times more free light chains than do lambda chain associated NMGs. In addition, the uninvolved kappa light chains in lambda chain associated lesions are, on average, present at twice the concentration of lambda light chains in kappa chain associated NMGs. Thus, if one were to use criteria specific for light chain type of lesions, the criterion “c” above should be amended to require that level of involved kappa light chain is >100 mg/L and lambda light chain >25 mg/L. The ratio of involved to uninvolved light chains should be 100 for kappa and 12.5 of lambda chain associated lesions (79).

Objective evidence for treating SMM based on myeloma defining condition is lacking and standard of care remains observation of SMM till symptomatic MM develops (78, 80, 81,). Evidence to support treating patients with symptomatic MM early rather than late has not been forthcoming. Prolonged overall survival has been reported with ASCT and also disputed by others (81–86,). There is general agreement that ASCT prolongs progression free survival but not overall survival (86).

Monitoring the Course of Light Chain MMs

SFLCA offers no advantage over urine examination in diagnosis of light chain MM. A case could be made for using SFLCA for monitoring the course of light chain MMs. The SFLCA provides a number that could make comparison of serial measurements meaningful. Using 24-h urine to calculate the change in production of SFLCs would be inconvenient, cumbersome, and not too accurate. The erroneously abnormal κ/λ ratios seen in IIg MM would be applicable to light chain MM as well (20, 87–89,). UIFE may be sufficient in monitoring light chain MM (47).

Light Chain Escape

In rare instances, subsequent to treatment, an IIg myeloma may recur as a light chain only disorder. The detection and monitoring of such patients may be facilitated by SFLCA (90,). However, the newly defined entity of LCPMM may provide a better explanation for the rare phenomenon described as light chain escape (30).

Minimal Residual Disease

As and when curative treatment for MM becomes available, determining minimal residual disease (MRD), may be warranted. For now, tests for ascertaining MRD are only relevant in randomized controlled trials (90, 91).

Measuring SFLCs

There is lack of standardization, and correlation amongst the various methods for measuring SFLC is inadequate. Reagents from a single vendor, when used on different platforms, provide disparate results. There are variations in normal populations by ethnicity in addition to variations induced by non-NMG disorders (92–99).

Measuring Monoclonal Serum Free Light Chains

An affinity purification and MALDI-TOF mass spectrometry assay has been described to detect monoclonal serum-free light chains; however, the assay is not quantitative and would offer no advantage over UIFE (31).

The QUIET method for measuring monoclonal SFLCs consists of ultrafiltration of serum using a nominal 50 kDa cut-off membrane, concentrating the filtrate and testing the concentrated specimen by immunofixation electrophoresis, using antisera to kappa and lambda light chains. Densitometry of the IFE gel provides quantity of monoclonal light chain (32,). It is speculated that the QUIET method could be used to detect minimal residual disease in patients with any type of MM by monitoring the monoclonal SFLCs, by using antisera specific to SFLCs (32).

In summary, serum and urine protein electrophoretic tests remain the gold standards for diagnosis and monitoring of monoclonal gammopathic disorders. The pathologist should review the EMR and issue a comprehensive report of various laboratory studies and a clinical correlation of the laboratory results. SFLCA adds minimal value and may be useful only in monitoring patients with light chain MM and perhaps LCPMM patients. Newer quantitative assays for monoclonal SFLC may add value in monitoring monoclonal light chain disorders and possibly in determining minimal residual disease in MM.

Nonstandard Abbreviations: SPEP, serum protein electrophoresis; SIFE, serum proteinimmunofixation electrophoresis; UPEP, urine protein electrophoresis; UIFE, urine protein immunofixation electrophoresis; CZE, capillary zone electrophoresis; SFLC, serum-free light chains; SFLCA, serum-free light chain assay; IMWG, international myeloma working group; NMG, neoplastic monoclonal gammopathies; MGUS, monoclonal gammopathy of undetermined significance; SMM, smoldering or asymptomatic myeloma; MM, plasma cell myeloma/multiple myeloma; MIg, monoclonal immunoglobulin; EMR, electronic medical record; LCPMM, light chain predominant multiple myeloma; QUIET, Quantification by Ultrafiltration and Immunofixation Electrophoresis Testing.

Author Contributions:All authors confirmed they have contributed to the intellectual content of this paper and have met the following 4 requirements: (a) significant contributions to the conception and design, acquisition of data, or analysis and interpretation of data; (b) drafting or revising the article for intellectual content; (c) final approval of the published article; and (d) agreement to be accountable for all aspects of the article thus ensuring that questions related to the accuracy or integrity of any part of the article are appropriately investigated and resolved.

Authors’ Disclosures or Potential Conflicts of Interest:Upon manuscript submission, all authors completed the author disclosure form. Disclosures and/or potential conflicts of interest:Employment or Leadership: None declared. Consultant or Advisory Role: G. Singh, Diazyme Inc., HealthTap. Stock Ownership: None declared. Honoraria: None declared. Research Funding: None declared. Expert Testimony: None declared. Patents: G. Singh, patent application pending for QUIET.

REFERENCES

1

Tate
JR.
 
The paraprotein – an enduring biomarker
.
Clin Biochem Rev
 
2019
;
40
:
5
22
.

2

Tate
JR
,
Graziani
MS
,
Moppee
P
,
Merlini
G
eds. Special Issue: Protein electrophoresis and serum free light chains in the diagnosis and monitoring of plasma cell disorders: Laboratory testing and current controversies. 2016:54 (June
2016
).

3

Le Carrer
D.
 Serum protein electrophoresis, immunofixation.
Illustrated interpretations. 2005 Ed.
 
Paris, France
:
FM-BIO Vanes
;
2005
.

4

Le Carrer
D
,
Boucraut
J.
 Urine protein electrophoresis, immunofixation.
Illustrated interpretations. 2005 edn.
 
Paris, France
:
FM-BIO Vanes
;
2005
.

5

Tate
J
,
Caldwell
G
,
Daly
J
,
Gillis
D
,
Jenkins
M
,
Jovanovich
S
, et al.  
Recommendations for standardized reporting of protein electrophoresis in Australia and New Zealand
.
Ann Clin Biochem
 
2012
;
49
:
242
56
.

6

Booth
RA
,
McCudden
CR
,
Balion
CM
,
Blasutig
IM
,
Bouhtiauy
I
,
Rodriguez-Capote
K
, et al.  
Candidate recommendations for protein electrophoresis reporting from the Canadian Society of Clinical chemists monoclonal gammopathy working group
.
Clin Biochem
 
2018
;
51
:
10
20
.

7

McCudden
CR
,
Booth
RA
,
Lin
DCC
,
McCurdy
A
,
Rupani
N
,
Kew
A.
 
Synoptic reporting for protein electrophoresis and immunofixation
.
Clin Biochem
 
2018
;
51
:
21
8
.

8

Genzen
JR
,
Murray
DL
,
Abel
G
,
Meng
QH
,
Baltaro
RJ
,
Rhoads
DD
, et al.  
Screening and diagnosis of monoclonal gammopathies. an international survey of laboratory practices
.
Arch Pathol Lab Med
 
2018
;
142
:
507
15
.

9

Wijeratne
N
,
Tate
JR
,
Wienholt
L
,
Molle
P.
 
Report of the survey conducted by RCPAQAP on current practice for paraprotein and serum free light chain mearurement and reporting. A need for harmonization
.
Clin Biochem Rev
 
2019
;
40
:
31
41
.

10

Keren
DF
,
Schroeder
L.
 
Challenges of measuring monoclonal proteins in serum
.
Clin Chem Lab Med
 
2016
;
54
:
947
61
.

11

Clavijo
A
,
Ryan
N
,
Xu
H
,
Singh
G.
 
Measurement of monoclonal immunoglobulin protein concentration in serum protein electrophoresis: comparison of automated vs manual/human readings
.
Lab Med
 
2020
;
51
:
252
8
.

12

Dimopoulos
M
,
Kyle
R
,
Fermand
JP
,
Rajkumar
SV
,
San Miguel
J
,
Chanan-Khan
A
, et al.  
Consensus recommendation for standard investigative workup: report of International Myeloma Workshop Consensus Panel
.
Blood
 
2011
;
117
:
4701
5
.

13

Rajkumar
SV
,
Dimopoulos
MA
,
Palumbo
A
,
Blade
J
,
Merlini
G
,
Mateos
MV
, et al.  
International Myeloma Working Group updated criteria for the diagnosis of multiple myeloma
.
Lancet Oncol
 
2014
;
15
:
e538
e548
.

14

Zia
H
,
Singh
G.
 
Optimization of utilization of serum protein analysis: Role of Electronic Medical Record in promoting consultation by pathologists
.
Am J Clin Pathol
 
2013
;
139
:
793
7
.

15

Kyle
RA
,
Therneau
TM
,
Rajkumar
SV
,
Larson
DR
,
Plevak
MF
,
Offord
JR
, et al.  
Prevalence of monoclonal gammopathy of undetermined significance
.
N Engl J Med
 
2006
;
354
:
1362
9
.

16

Kyle
RA
,
Larson
DR
,
Therneau
TM
,
Dispenzieri
A
,
Kumar
S
,
Cerhan
JR
,
Rajkumar
SV.
 
Long-term fellow-up of monoclonal gammopathy of undetermined significance
.
N Engl J Med
 
2018
;
378
:
241
9
.

17

Kyle
RA
,
Remstein
ED
,
Therneau
TM
,
Dispenzieri
A
,
Kurtin
PJ
,
Hodnefield
JM
, et al.  
Clinical course and prognosis of smoldering (asymptomatic) multiple myeloma
.
N Engl J Med
 
2007
;
356
:
2582
90
.

18

Drouet
E
,
Chapuis-Cellier
C
,
Bosshard
S
,
Verniol
C
,
Niveleau
A
,
Touraine
JL
,
Garnier
JL.
 
Oligo-monoclonal immunoglobulins frequently develop during concurrent cytomegalovirus (CMV) and Epstein-Barr virus (EBV) infections in patients after renal transplantation
.
Clin Exp Immunol
 
1999
;
118
:
465
72
.

19

Tang
F
,
Malek
E
,
Math
S
,
Schmotzer
CL
,
Beck
RC.
 
Interference of therapeutic monoclonal antibodies with routine serum protein electrophoresis and immunofixation in patients with myeloma. Frequency and duration of detection of Daratumumab and Elotusumab
.
Am J Clin Pathol
 
2018
;
150
:
121
9
.

20

Singh
G.
 
Serum free light chain assay and κ/λ ratio: performance in patients with monoclonal gammopathy-high false negative rate for κ/λ Ratio
.
J Clin Med Res
 
2017
;
9
:
46
57
.

21

Heaton
C
,
Vyas
SG
,
Singh
G.
 
Audit of use and overuse of serum protein immunofixation electrophoresis and serum free light chain assay in tertiary healthcare. A case for algorithmic testing to optimize laboratory utilization
.
Am J Clin Pathol
 
2016
;
145
:
531
7
.

22

Chen
C
,
Chen
P
,
Fang
X.
 
The FLC dimer with lambda type may false-migrate to the position of “albumin” band by urine protein electrophoresis on Sebia gel-based detection system
.
J Clin Lab Anal
 
2019
;
33
:
e22658
.

23

Murata
K
,
McCash
SI
,
Carroll
B
,
Lesokhin
AM
,
Hassoun
H
,
Lendvai
N
, et al.  
Treatment of multiple myeloma with monoclonal antibodies and the dilemma of false positive M-spikes in peripheral blood
.
Clin Biochem
 
2018
;
51
:
66
71
.

24

Finn
WG
,
Gulbranson
R
,
Fisher
S
,
Rae Sample
L
,
Shalhoub
R
,
Hedstrom
D
,
Keren
DF.
 
Detection of polyclonal increase in immunoglobulin G4 subclass by distinct patterns on capillary serum protein electrophoresis. Diagnostic pitfalls and clinical observations in a study of 303 cases
.
Am J Clin Pathol
 
2016
;
146
:
303
11
.

25

Vyas
SG
,
Singh
G.
 
Location of monoclonal peak as a tool in checking specimen integrity
.
Pract Lab Med
 
2017
;
8
:
49
51
.

26

Vyas
SG
,
Singh
G.
 
Prospective identification of oligoclonal/abnormal band of the same immunoglobulin type as the malignant clone by differential location of the M-spike and oligoclonal band
.
J Clin Med Res
 
2017
;
9
:
826
30
.

27

Singh
G.
 
Serum free light chain assay and κ/λ ratio: performance in patients without monoclonal gammopathies – high false positive rate
.
Am J Clin Pathol
 
2016
;
146
:
207
14
.

28

Hall
SL
,
Tate
J
,
Gill
D
,
Mollee
P.
 
Significance of abnormal protein bands in patients with multiple myeloma following autologous stem cell transplantation
.
Clin Biochem Rev
 
2009
;
30
:
113
8
.

29

Singh
G.
 
Concentration of serum free light chains in kappa and lambda lesions in light-chain myelomas
.
Lab Med
 
2019
;
50
:
189
93
.

30

Singh
G, Xu H.
 
Light chain predominant intact immunoglobulin monoclonal gammopathy disorders
.
Lab Med, in Press
 
2020
;
XX
:
1
7
.

31

Sepiashvili
L
,
Kohlhagen
MC
,
Snyder
MR
,
Willrich
MAV
,
Mills
JR
,
Dispenzieri
A
,
Murray
DL.
 
Direct detection of monoclonal free light chains in serum by use of immunoenrichment-coupled MALDI-TOF mass spectrometry
.
Clin Chem
 
2019
;
65
:
1015
22
.

32

Singh
G
,
Bollag
R.
 
Quantification by Ultrafiltration and Immunofixation Electrophoresis Testing (QUIET) for monoclonal serum free light chains
.
Lab Med: In Press as
. DOI: 10.1093/labmed/lmaa012.

33

Katzmann
JA
,
Willrich
MAV
,
Kohlhagen
MC
,
Kyle
RA
,
Murray
DL
,
Snyder
MR
, et al.  
Monitoring IgA multiple myeloma: immunoglobulin heavy/light chain assays
.
Clin Chem
 
2015
;
61
:
360
7
.

34

Beaumont-Epinette
M-P
,
Moreau
C
,
Besnard
S
,
Latute
F
,
Collet
N
,
Sebillot
M
, et al.  
Heavy/light chain specific immunoglobulin ratio provides no additional information than serum protein electrophoresis and immunofixation for the diagnosis and follow-up of intact immunoglobulin multiple myeloma patients
.
Pathol Biol
 
2015
;
63
:
215
21
.

35

Kyle
RA.
 
Criteria for the classification of monoclonal gammopathies, multiple myeloma and related disorders: a report of the international myeloma working group
.
British J Haematol
 
2003
;
121
:
749
57
.

36

Katzman
JA
,
Dispenzieri
A
,
Kyle
RA
,
Snyder
MR
,
Plevak
MF
,
Larson
DR
, et al.  
Elimination of the need for urine studies in the screening algorithm for monoclonal gammopathy by using serum immunofixation and free light chain assay
.
Mayo Clin Proc
 
2006
;
81
:
1575
8
.

37

Wood
PB
,
McElroy
YG
,
Stone
MJ.
 
Comparison of serum immunofixation electrophoresis and free light chain assay in detection of monoclonal gammopathies
.
Clin Lymphoma Myeloma Leuk
 
2010
;
10
:
278
80
.

38

Boer
K
,
Deufel
T.
 
Quantitation of serum free light chains does not compensate for serum immunofixation only when screening for monoclonal gammopathies
.
Clin Chem Lab Med
 
2009
;
47
:
1109
15
.

39

Levinson
SS.
 
Complementarily of urine analysis and serum free light chain assay for assessing responses to treatment response. Illustrated by three case examples
.
Clin Chim Acta
 
2011
;
412
:
2206
10
.

40

Shaheen
SP
,
Levinson
SS.
 
Serum free light assay may miss monoclonal light chains that urine immunofixation electrophoresis would detect
.
Clin Chim Acta
 
2009
;
406
:
162
6
.

41

Rao
M
,
Yu
WW
,
Chan
J
,
Patel
K
,
Comenzo
R
,
Lamont
JL
,
Ip
S
,
Lau
J.
 
Comparative effectiveness reviews, no. 73 Serum free light chain analysis for the diagnosis, management and prognosis of plasma cell dyscrasias
.
Rockville (MD
):
Agency for Healthcare Research and Quality (US
);
2012
Aug. Report No.: 12-EHC102-EF

42

Attaelmannan
M
,
Levinson
SS.
 
Understanding and identifying monoclonal gammopathies
.
Clin Chem
 
2000
;
46
:
1230
8
.

43

Lee
WS
,
Singh
G.
 
Serum free light chains in neoplastic monoclonal gammopathies: Relative under-detection of lambda dominant kappa/lambda ratio and underproduction of free lambda light chains, as compared to kappa light chains in patients with neoplastic monoclonal gammopathies
.
J Clin Med Res
 
2018
;
7
:
562
9
.

44

Kumar
S
,
Larson
DR
,
Dispenzieri
A
,
Therneau
TM
,
Murray
DL
,
Leif Bergsagel
P
, et al.  
Polyclonal serum free light chain elevation is associated with increased risk of monoclonal gammopathies
.
Blood Cancer J
 
2019
;
49–54
:https://doi.org/10.1038/s41408-019-0210-z.

45

Singh
G.
 
Oligoclonal pattern/abnormal protein bands in post-treatment plasma cell myeloma patients: Implications for electrophoresis and serum free light chain assay results
.
J Clin Med Res
 
2017
;
9
:
671
9
.

46

Brebner
JA
,
Stockley
RA.
 
Polyclonal free light chains: a biomarker of inflammatory disease or treatment target?
 
F1000 Med Rep
 
2013
;
5
:
4
.

47

Brigden
ML
,
Neal
ED
,
Mcneely
MDD
,
Hoag
GN.
 
The optimum urine collection for the detection and monitoring of Bence Jones proteinuria
.
Am J Clin Pathol
 
1990
;
93
:
689
93
.

48

Rajkumar
SV
,
Kyle
RA
,
Therneau
YM
,
Therneau
TM
,
Melton
J
,
Bradwell
AR
, et al.  
Serum free light chain ratio is an independent risk factor for progression in monoclonal gammopathy of undetermined significance
.
Blood
 
2005
;
106
:
812
7
.

49

Thompson
CA
,
Maurer
MJ
,
Katzmann
JA
,
Ansell
SM
,
Habermann
TM
,
Macon
WR
, et al.  
Elevated serum free light chains are associated with inferior event free survival in Hodgkin lymphoma
.
Am J Hematol
 
2011
;
86
:
998
1000
.

50

Anandram
S
,
Assi
LK
,
Lovatt
T
,
Parkes
J
,
Taylor
J
,
Macwhannell
A
, et al.  
Elevated, combined serum free light chain levels and increased mortality: a 5-year follow-up UK study
.
J Clin Pathol
 
2012
;
65
:
1036
42
.

51

Dispenzieri
A
,
Katzmann
JA
,
Kyle
RA
,
Larson
DR
,
Therneau
TM
,
Colby
CL
, et al.  
Use of nonclonal serum immunoglobulin free light chains to predict overall survival in general population
.
Mayo Clin Proc
 
2012
;
87
:
517
23
.

52

Moen
IW
,
Bergholdt
HKM
,
Mandrup-Poulsen
T
,
Nordestgaard
BG
,
Ellervik
C.
 
Increased plasma ferritin concentration and low-grade inflammation-A Mendelian Randomization Study
.
Clin Chem
 
2018
;
64
:
374
85
.

53

Burmeister
A
,
Assi
LK
,
Ferro
CJ
,
Hughes
RG
,
Barnett
AH
,
Bellary
S
, et al.  
The relationship between high-sensitivity CRP and polyclonal free light chains as markers of inflammation in chronic disease
.
Int J Lab Hematol
 
2014
;
36
:
415
24
.

54

Witzig
TE
,
Maurer
MI
,
Haberman
TM
,
Link
BK
,
Micallef
INM
,
Nowakowski
GS
, et al.  
Elevated monoclonal and polyclonal serum immunoglobulin free light chain (FLC) as prognostic factors in B- and T-cell non-Hodgkin lymphoma
.
Am J Hematol
 
2014
;
89
:
1116
20
.

55

Bellary
S
,
Faint
JM
,
Assi
LK
,
Hutchison
CA
,
Harding
SJ
,
Raymond
NT
,
Barnett
AH.
 
Elevated serum free light chains predict cardiovascular events in type 2 diabetes
.
Diabetes Care
 
2014
;
37
:
2028
30
.

56

Fulks
M
,
Stout
RL
,
Dolan
VF.
 
Albumin and all-cause mortality risk in insurance applicants
.
J Insur Med
 
2010
;
42
:
11
7
.

57

Li
Y
,
Zhong
X
,
Cheng
G
,
Zhao
C
,
Zhang
L
,
Hong
Y
, et al.  
Hs-CRP and all-cause, cardiovascular, and cancer mortality risk: a meta-analysis
.
Atherosclerosis
 
2017
;
259
:
75
82
.

58

Chen
CM
,
Lee
M
,
Yang
YH
,
Huang
SS
,
Lin
CH.
 
Association between clinical and laboratory markers and 5-year mortality among patients with
.
Sci Rep
 
2019
;
9
:
11521
30
.

59

Uljon
SN
,
Richardson
PG
,
Schur
PH
,
Anderson
KC
,
Tanasijevic
MJ
,
Lindeman
NI.
 
Serial serum free light chain measurements do not detect changes in disease status earlier than electrophoretic M-spike measurements in patients with intact immunoglobulin myeloma
.
Clin Chim Acta
 
2011
;
412
:
562
8
.

60

Rhee
F
,
Bolejack
V
,
Hollmig
K
,
Pineda-Roman
M
,
Anaissie
E
,
Epstein
J
, et al.  
High serum free light chain levels and their rapid reduction in response to therapy define an aggressive multiple myeloma subtype with poor prognosis
.
Blood
 
2007
;
110
:
827
32
.

61

Li
W
,
Zhou
JZ
,
Chang
HR
,
Dai
L
,
Zhu
Z
,
Feng
V
, et al.  
Comparison of SPE, IFE and FLC in monitoring patients with multiple myeloma after autologous stem cell transplantation
.
Cell Biochem Biophys
 
2015
;
73
:
643
7
.

62

de Larrea
CF
,
Cibeira
MT
,
Elena
M
,
Arostegui
JI
,
Rosiñol
L
,
Rovira
M
, et al.  
Abnormal serum free light chain ratio in patients with multiple myeloma in complete remission has strong association with the presence of oligoclonal bands: implications for stringent complete remission definition
.
Blood
 
2009
;
114
:
4954
6
.

63

Barley
K
,
Tindle
S
,
Bagiella
E
,
Jagannath
S
,
Chari
A.
 
Serum free light chain assessment early after stem cell transplantation as a prognostic factor in multiple myeloma
.
Clin Lymphoma Myeloma Leuk
 
2015
;
15
:
541
5
.

64

Fujisawa
M
,
Seike
K
,
Fukumoto
K
,
Suehara
Y
,
Fukaya
M
,
Sugihara
H
, et al.  
Oligoclonal bands in patients with multiple myeloma: its emergence per se could not be translated to improved survival
.
Cancer Sci
 
2014
;
105
:
1442
6
.

65

Shaw
GR.
 
Nonsecretory plasma cell myeloma-becoming even more rare with serum free light-chain assay: a brief review
.
Arch Pathol Lab Med
 
2006
;
130
:
1212
5
.

66

Kumar
SK
,
Dispenzieri
A
,
Lacy
MQ
,
Hayman
SR
,
Buadi
FK
,
Zeldenrust
SR
, et al.  
Changes in serum-free light chains rather than intact monoclonal immunoglobulin levels predict outcome following therapy in primary amyloidosis
.
Am J Hematol
 
2011
;
86
:
251
5
.

67

Gatt
ME
,
Kaplan
B
,
Yogev
D
,
Slyusarevsky
E
,
Pogrebijski
G
,
Golderman
S
, et al.  
The use of serum free light chain dimerization patterns assist in the diagnosis of AL amyloidosis
.
Br J Haematol
 
2018
;
182
:
86
92
.

68

Nguyen
VP
,
Rosenberg
A
,
Mendelson
LM
,
Comenzo
RL
,
Varga
C
,
Sanchorawala
V.
 
Outcomes of patients with AL amyloidosis and low serum free light chain levels at diagnosis
.
Amyloid
 
2018
;
25
:
156
9
.

69

Bhole
MV
,
Sadler
R
,
Ramasamy
K.
 
Serum-free light-chain assay: clinical utility and limitations
.
Ann Clin Biochem
 
2014
;
51
:
528
42
.

70

Abbi
KKS
,
Silverman
M
,
Farooq
F
,
Tricot
A
,
Dozeman
L
,
Nadiminti
K
, et al.  
Potential pitfalls of serum free light analysis to assess treatment response for multiple myeloma
.
Br J Haematol
 
2016
;
174
:
536
40
.

71

Moustafa
MA
,
Rajkumar
SV
,
Dispenzieri
A
,
Gertz
MA
,
Lacy
MQ
,
Buadi
FK
, et al.  
Utility of serum free light chain measurements in multiple myeloma patient not achieving complete response to therapy
.
Leukemia
 
2015
;
29
:
2033
8
.

72

Martinez-Lopez
J
,
Paiva
B
,
Lopez-Anglada
L
,
Mateos
MV
,
Cedena
T
,
Vidríales
MB
, et al.  
Critical analysis of the stringent complete response in multiple myeloma: contribution of sFLC and bone marrow clonality
.
Blood
 
2015
;
126
:
858
62
.

73

Lopez-Anglada
L
,
Cueto-Felgueroso
C
,
Rosiñol
L
,
Oriol
A
,
Teruel
AI
,
Lopez de la Guia
A
, et al.  
Prognostic utility of serum free light chain ratios and heavy-light chain ratios in multiple myeloma in three PETHEMA/GEM phase III clinical trials
.
PLoS One
 
2018
;
13
:
e0203392
.

74

Chee
CE
,
Kumar
S
,
Larson
DR
,
Kyle
RA
,
Dispenzieri
A
,
Gertz
MA
, et al.  
the importance of bone marrow examination in determining complete response to therapy in patients with multiple myeloma
.
Blood
 
2009
;
114
:
2617
20
.

75

Paiva
B
,
Martinez-Lopez
J
,
Vidriales
M
,
Mateos
M
,
Montalban
M
,
Fernandez-Redondo
E
, et al.  
Comparison of immunofixation, serum free light chain and immunophenotyping for response evaluation and prognostication in multiple myeloma
.
J Clin Oncol
 
2011
;
29
:
1627
33
.

76

Larsen
JT
,
Kumar
SK
,
Dispenzieri
A
,
Kyle
JA
,
Katzmann
JA
,
Rajkumar
SV.
 
Serum free light chain ratio as a biomarker for high-risk smoldering multiple myeloma
.
Leukemia
 
2013
;
27
:
941
9466
.

77

Rajkumar
SV.
Updated diagnostic criteria and staging system for multiple myeloma.
2016
ASCO Education Book
e418
e422
.

78

Caers
J
,
Fernández de Larrea
C
,
Leleu
X
,
Heusschen
R
,
Zojer
N
,
Decaux
O
, et al.  
The changing landscape of smoldering multiple myeloma: a European perspective
.
Oncologist
 
2016
;
21
:
333
42
.

79

Lee
WS
,
Singh
G.
 
Serum free light chain assay in monoclonal gammopathies manifestations
.
Lab Med
 
2019
;
50
:
381
9
.

80

Rajkumar
SV
,
Landgren
O
,
Mateos
M-V.
 
Smoldering multiple myeloma
.
Blood
 
2015
;
125
:
3069
75
.

81

Kapoor
P
,
Rajkumar
SV.
 
Smoldering multiple myeloma: to treat or not to treat
.
Cancer J
 
2019
;
25
:
65
71
.

82

Goldschmidt
N
,
Zamir
L
,
Poperno
A
,
Kahan
NR
,
Paltiel
O.
 
Presenting signs of multiple myeloma and effect of diagnostic delay on prognosis
.
J Am Board Fam Med
 
2016
;
29
:
702
9
.

83

Thoennissen
GB
,
Görlich
D
,
Bacher
U
,
Aufenberg
T
,
Hüsken
A-C
,
Hansmeier
AA
, et al.  
Autologous stem cell transplantation in multiple myeloma in the era of novel induction: A retrospective single-center analysis
.
Acta Haematol
 
2017
;
137
:
163
72
.

84

Perez
R
,
Duran
MS
,
Mayans
J
,
Soler
A
,
Castillo
I
,
Jurado
M
, et al.  
Clinical features and survival of 338 multiple myeloma patients treated with hematopoietic stem cell transplantation or conventional chemotherapy
.
Eur J Haematol
 
2016
;
96
:
417
24
.

85

Dhakal
B
,
Szabo
A
,
Chhabra
S
,
Hamadani
M
,
D’Souza
A
,
Usmani
SZ
, et al.  
Autologous transplantation for newly diagnosed multiple myeloma in the era of novel agent induction: a systematic review and meta-analysis
.
JAMA Oncol
 
2018
;
4
:
343
50
.

86

Attal
M
,
Lauwers-Cances
V
,
Hulin
C
,
Leleu
X
,
Caillot
D
,
Escoffre
M
, et al.  
Lenalidomide, bortezomib, and dexamethasone with transplantation for myeloma
.
N Engl J Med
 
2017
;
376
:
1311
20
.

87

Zamora-Ortiz
G
,
Velazquez-Sanchez-de-Cima
S
,
Hernandes-Reyes
J
,
Martagón-Herrera
NA
,
Ruiz-Argüelles
A
,
Ruiz-Delgado
GJ
,
Ruiz-Argüelles
GJ.
 
Poor performance of the total kappa/lambda light chain quantification in the diagnosis and fellow-up of patients with multiple myeloma
.
Rev Invest Clin
 
2014
;
66
:
314
8
.

88

Dejoie
T
,
Corre
J
,
Caillon
H
,
Hulin
C
,
Perrot
A
,
Caillot
D
, et al.  
Serum free light chains, not urine specimens, should be used to evaluate response in light-chain multiple myeloma
.
Blood
 
2016
;
128
:
2941
8
.

89

Brioli
A
,
Giles
H
,
Pawlyn
C
,
Campbell
JP
,
Kaiser
MF
,
Melchor
L
, et al.  
Serum free immunoglobulin light chain evaluation as a marker of impact from intraclonal heterogeneity on myeloma outcome
.
Blood
 
2014
;
123
:
3414
9
.

90

Innao
V
,
Allegra
A
,
Russo
S
,
Gerace
D
,
Vaddinelli
D
,
Alonci
A
, et al.  
Standardisation of minimal residual disease in multiple myeloma
.
Eur J Cancer Care
 
2017
;
26
:
e12732
.

91

Kumar
S
,
Paiva
B
,
Anderson
KC
,
Durie
B
,
Landgren
O
,
Moreau
P
, et al.  
International myeloma working group consensus for response and minimal residual disease assessment in multiple myeloma
.
Lancet Oncol
 
2016
;
17
:
e328
e346
.

92

Fleming
CKA
,
Swarttouw
T
,
de Kat Angelino
CM
,
Jacobs
JFM
,
Russcher
H.
 
Method comparison of four clinically available assays for serum free light chain analysis
.
Clin Chem Lab Med
 
2019
;
58
:
85
94
.

93

Jacobs
JF
,
Tate
JR
,
Merlini
G.
 
Is accuracy of serum free light chain measurement achievable?
 
Clin Chem Lab Med
 
2016
;
54
:
1021
30
.

94

Kennard
A
,
Hawley
C
,
Tate
J
,
Klingberg
S
,
Pretorius
C
,
Hutchison
C
,
Mollee
P.
 
Comparison of FreelieTM and N Latex serum free light chain assay in subjects with end stage kidney disease on haemodilysis
.
Clin Chem Lab Med
 
2016
;
54
:
1045
52
.

95

Katzmann
JA
,
Clark
RJ
,
Abraham
RS
,
Bryant
S
,
Lymp
JF
,
Bradwell
AR
,
Kyle
RA.
 
Serum reference intervals and diagnostic ranges for free κ and free λ immunoglobulin light chains: relative sensitivity for detection of monoclonal light chains
.
Clin Chem
 
2002
;
48
:
1437
44
.

96

Cotten
SW
,
Shajani-Yi
Z
,
Cervinski
MA
,
Voorhees
T
,
Tuchman
SA
,
Korpi-Steiner
N.
 
Reference intervals and diagnostic ranges for serum free κ and free λ immunoglobulin light chains vary by instrument platform: implications for classification of patient results in multi-center study
.
Clin Biochem
 
2018
;
58
:
100
7
.

97

Messain
A-S
,
De Sloovere
MMW
,
Claus
P-E
,
Vercammen
M
,
Van Hoovels
L
,
Heylen
O
, et al.  
Performance evaluation of serum free light chain analysis. Nephelometry vs turbidimetry, monoclonal vs polyclonal reagents
.
Am J Clin Pathol
 
2017
;
147
:
611
22
.

98

Liang
YF
,
Chen
VVM
,
Wang
QT
,
Zhai
Y
,
Yang
Y
,
Liu
J.
 
Establishment and validation of serum free light chain reference intervals in an ethnic Chinese population
.
Clin Lab
 
2014
;
60
:
193
8
.

99

Galvani
L
,
Flanagan
J
,
Sargazi
M
,
Neithercut
WD.
 
Annals express: validation of serum free light chain reference ranges in primary care patients
.
Ann Clin Biochem
 
2016
;
53
:
399
404
.

Nonstandard Abbreviations

     
  • SPEP

    serum protein electrophoresis

  •  
  • SIFE

    serum protein immunofixation electrophoresis

  •  
  • UPEP

    urine protein electrophoresis

  •  
  • UIFE

    urine protein immunofixation electrophoresis

  •  
  • CZE

    capillary zone electrophoresis

  •  
  • SFLC

    serum-free light chains

  •  
  • SFLCA

    serum-free light chain assay

  •  
  • IMWG

    international myeloma working group

  •  
  • NMG

    neoplastic monoclonal gammopathies

  •  
  • MGUS

    monoclonal gammopathy of undetermined significance

  •  
  • SMM

    smoldering or asymptomatic myeloma

  •  
  • MM

    plasma cell myeloma/multiple myeloma

  •  
  • MIg

    monoclonal immunoglobulin

  •  
  • EMR

    electronic medical record

  •  
  • LCPMM

    light chain predominant multiple myeloma

  •  
  • QUIET

    Quantification by Ultrafiltration and Immunofixation Electrophoresis Testing

This article is published and distributed under the terms of the Oxford University Press, Standard Journals Publication Model (https://dbpia.nl.go.kr/journals/pages/open_access/funder_policies/chorus/standard_publication_model)