Since the mid-1990s, the multiple myeloma treatment landscape has evolved considerably, which has led to improved patient outcomes and prolonged survival. In addition to discovering new, targeted agents or treatment regimens, the identification and validation of biomarkers has the potential to further improve patient outcomes. The International Staging System relies on a number of biochemical parameters to stratify patients into risk categories. Other biologically relevant markers that are indicative of inherited genetic variation (e.g., single-nucleotide polymorphisms) or tumor-acquired genetic events (e.g., chromosomal translocations or mutations) have been studied for their prognostic potential. In patients with high-risk cytogenetics, plasma cells (PC) undergo genetic shifts over time, which may partially explain why high-risk patients relapse and are so difficult to treat. Although novel agents have improved treatment outcomes, identification of markers that will enable clinicians to determine which treatment is most appropriate for high-risk patients following initial diagnosis represents an exciting frontier in the clinical management of multiple myeloma. Biomarkers based on quantitating PCs or factors that are secreted from them (e.g., serum free light chain) may also help to risk-stratify patients with asymptomatic multiple myeloma. Eventually, identification of novel biomarkers may lead to the creation of personalized treatment regimens that are optimized to target clonal PCs that express a specific oncogenomic profile. Although the future is exciting, validation will be necessary before these biologic and molecular beacons can inform decision-making processes in a routine clinical setting. Clin Cancer Res; 20(4); 804–13. ©2013 AACR.

Multiple myeloma is a hematologic disease that is characterized by the proliferation of abnormal bone marrow plasma cells (BMPC) and immunoglobulin or light chain overproduction, with evidence of end-organ damage. Before 1997, the median survival of patients with newly diagnosed multiple myeloma was approximately 2.5 years (1). From 1997 to 2006, the use of high-dose anti–multiple myeloma therapy, stem cell transplantation, and novel agents increased the median overall survival (OS) to nearly 4 years, a 50% improvement (1). In some patients who are currently receiving such agents, improved relapse-free survival has increased to >10 years (2), raising the potential for long-term disease control and care.

Although the multiple myeloma treatment landscape has improved since the mid-1990s, the intraclonal heterogeneity (ICH) of malignant plasma cells (PC), the interaction of PCs with host factors, and the bone marrow microenvironment contribute to disease progression and the molecular evolution of the disease according to Darwinian principles (3, 4). Taken together, these factors contribute to the generation of treatment-resistant PCs, which ultimately leads to relapsed and refractory myeloma, the disease stage that is frequently used for drug development. However, as the nature of this disease stage continues to change as a consequence of patients receiving multiple lines of therapy, it is essential that we develop novel approaches to drug development.

If patients are to overcome treatment resistance, one approach that may improve clinical outcomes is to select treatments that are well matched to a patient's molecularly defined disease subtype (5). Another promising approach is to identify new biomarkers and tools that can identify onset or worsening of disease. According to the National Cancer Institute (NCI), a biomarker is a molecule that is found in blood and other body fluids or tissues that can serve as an indicator for a normal or an abnormal process, condition, or disease (6). Herein, we review biomarkers that have been used in multiple myeloma that meet the NCI's definition of a biomarker [e.g., monoclonal (M) protein]; we will also discuss molecular markers and tools that have the potential to dramatically alter how clinicians diagnose, stage, and treat patients with multiple myeloma or asymptomatic “early” multiple myeloma.

The emergence of clinical biomarkers in multiple myeloma

In the 1960s, multiple myeloma researchers began to identify biomarkers that were independent predictors of survival, including hemoglobin, serum calcium, serum creatinine, and bone lesion severity (7, 8). In 1975, Durie and Salmon (9) introduced a staging system that used M protein, hemoglobin, calcium, and a number of bone lesions to predict multiple myeloma cell tumor burden. In the 1980s, serum β2-microglobulin (β2M) was found to be a simple, yet reliable prognostic marker for disease staging; serum β2M also enabled clinicians to predict a patient's likelihood of survival (10, 11). Subsequently, albumin (12), C-reactive protein (13), and BMPC proliferation indices (14, 15) were found to be reliable prognostic factors, but they did not come into general use. In 2005, an international consortium of researchers used serum β2M and serum albumin to create the International Staging System (ISS), which enabled clinicians to stage patients and predict their long-term prognosis (16). This has been further refined by combining FISH data with ISS (17, 18).

Other types of biomarkers [e.g., serum free light chain (sFLC) ratio and cytogenetic markers] are also beginning to provide prognostic information on multiple myeloma and patients with asymptomatic disease (18–20). There have been significant developments in the diagnostic platforms that are available to assess the molecular features of multiple myeloma that provide extensive novel information as well as new diagnostic tests. The current emphasis is on using multiparallel genome-wide approaches, which yield huge amounts of data and offer the advantage of being able to multiplex genomes from many patients in a single run, which can reduce the cost per test. However, these technologies also raise significant issues with regard to the generation of complex datasets and how they can be reliably analyzed. These technologies will need to be robust and applicable if they are to be widely used in a routine setting. It is also appropriate to ask whether we should take a genome-wide approach to biomarker development or whether we should focus on a smaller number of “multiple myeloma relevant” markers that can be analyzed using simpler and potentially more robust techniques.

Cytogenetic biomarkers in multiple myeloma

Changes at the DNA level determine how a cancer cell behaves. If we can understand these molecular events, we should be able to predict the behavior of cancer. Chromosomal translocations are primary genetic events that occur early in the disease and are seen in roughly 50% of patients with multiple myeloma. As a consequence of the translocation events, there is overexpression of partner genes, leading to abnormal clonal PC behavior and differing clinical outcomes (17, 21–27). For example, 4-year progression-free survival (PFS) and OS rates for patients with the t(4;14) have been estimated to be 11% and 35% versus PFS and OS rates of 32% and 60% for patients without the t(4;14), respectively (18).

Other chromosomal translocations have been investigated for their prognostic potential. The t(14;16) is present in <5% of patients with newly diagnosed multiple myeloma and defines a group of patients who often have poor survival (17, 18, 28, 29). Although the t(14;16) is relatively rare, the MAF gene is overexpressed in up to 30% of patients with multiple myeloma, defining a greater number of patients with a unifying biologic feature (4). The t(11;14) is observed in approximately 20% of patients and is associated with a favorable prognosis, higher rates of CD20 expression, lymphoplasmacytic morphology, hyposecretory disease, λ-light chain usage, and nuclear cyclin D1 (CCND1) expression and dysregulation (18, 21, 23, 30–32).

Hyperdiploidy of the odd-numbered chromosomes is the other major primary genetic event in multiple myeloma that defines the other 50% of multiple myeloma cases (Table 1; refs. 4, 33–36). Although the exact cause of hyperdiploidy is unknown, it seems to define a group of patients with a more favorable prognosis (37). In addition to chromosomal translocations and hyperdiploidy, inherited genetic variation is an important factor in the etiology of multiple myeloma. Molecular epidemiology techniques have shown that multiple myeloma is associated with single-nucleotide polymorphisms (SNP) at the chromosomal regions 2p, 3p, 6p, 7p, 17p, and 11q (4, 34, 35). However, at this point, no prognostic value has been seen in association with these variables. Inherited variation can also affect response to chemotherapy and the side-effect profile of a drug. To date, very few studies have been carried out to test this hypothesis, and they have not as yet defined relevant markers that can be used in this way in multiple myeloma (38).

Table 1.

Inherited variation and primary genetic events in multiple myeloma

Inherited variation
SNPs
ChromosomeGenesPrimary toolsReferences
 2p23.3 DTNB and DNMT3A GWAS (34, 35) 
 3p22.1 ULK4 and TRAK1   
 7p15.3 DNAH11 and CDCA7L   
Primary genetic events 
IGH@ translocations    
Translocation Genes Primary tools References 
 t(4;14) at 4p16.3 FGFR3 and MMSET Karyotype analysis (cytogenetics), FISH, GEP, GWAS, RT-PCR, or sequencing (25) 
 t(6;14) at 6p21 CCND3  (21, 23) 
 t(11;14)(p21;q11) CCND1  (21, 23, 27) 
 t(14;16)(q32;q23) MAF  (24) 
 t(14;20)(q32;q12) MAFB  (17, 26) 
Hyperdiploidy (chromosomal trisomy)    
Chromosome Genes Primary tools References 
 3, 5, 7, 9, 11, 15, 19, and 21  GEP and FISH (41) 
Inherited variation
SNPs
ChromosomeGenesPrimary toolsReferences
 2p23.3 DTNB and DNMT3A GWAS (34, 35) 
 3p22.1 ULK4 and TRAK1   
 7p15.3 DNAH11 and CDCA7L   
Primary genetic events 
IGH@ translocations    
Translocation Genes Primary tools References 
 t(4;14) at 4p16.3 FGFR3 and MMSET Karyotype analysis (cytogenetics), FISH, GEP, GWAS, RT-PCR, or sequencing (25) 
 t(6;14) at 6p21 CCND3  (21, 23) 
 t(11;14)(p21;q11) CCND1  (21, 23, 27) 
 t(14;16)(q32;q23) MAF  (24) 
 t(14;20)(q32;q12) MAFB  (17, 26) 
Hyperdiploidy (chromosomal trisomy)    
Chromosome Genes Primary tools References 
 3, 5, 7, 9, 11, 15, 19, and 21  GEP and FISH (41) 

Abbreviations: GEP, global gene expression profiling; GWAS, genome-wide association study.

Secondary genetic events, which are important in disease progression, also affect various molecular pathways and contribute to the biologic heterogeneity of PCs (Table 2). In this context, prognostically important copy number abnormalities (including gains or deletions of whole chromosomes or interstitial copy number gains) have been identified using SNP mapping arrays (4, 18, 33, 39). Clinically relevant regions that have been identified using this approach include 1q+, 1p32−, 1p11−, and 17p−. The International Myeloma Working Group (IMWG) has recommended assessing these specific cytogenetic lesions at disease presentation, as they may have a prognostic value that is clinically relevant. The study of copy number change has also improved our understanding of the biology of multiple myeloma, leading to the identification of 16q, a WW-domain–containing oxidoreductase gene (WWOX), CYLD (a negative regulator of the NF-κβ pathway), 11q− (BIRC2 and BIRC3), and 14q (TRAF3) as being relevant recurrent events (40, 41). These findings reinforce the importance of using therapeutic approaches to target the NF-κβ pathway.

Table 2.

Secondary genetic events in multiple myeloma

ChromosomesGenesToolsReferences
Secondary genetic events 
Secondary translocations    
 t(8;14) MYC FISH (75) 
Gains 
 1q CKS1B and ANP32E GEP (41) 
 12p LTBR   
 17q  Comparative genomic hybridization (76) 
Deletions 
 1p CDKN2C, FAF1, and FAM46C GEP, GM, and FISH (41, 77) 
 6q   (41) 
 8p TRAIL-R1 and TRAIL-R2  (41, 78) 
 11q BIRC2 and BIRC3  (79, 80) 
 13 RB1 and DIS3  (41) 
 14q TRAF3  (79, 80) 
 16q CYLD and WWOX  (40, 41) 
 17p TP53  (41, 81) 
Epigenetic events 
Global hypomethylation (MGUS to multiple myeloma) and gene-specific hypermethylation (multiple myeloma to PC leukemia) Genome-wide methylation arrays (49)  
ChromosomesGenesToolsReferences
Secondary genetic events 
Secondary translocations    
 t(8;14) MYC FISH (75) 
Gains 
 1q CKS1B and ANP32E GEP (41) 
 12p LTBR   
 17q  Comparative genomic hybridization (76) 
Deletions 
 1p CDKN2C, FAF1, and FAM46C GEP, GM, and FISH (41, 77) 
 6q   (41) 
 8p TRAIL-R1 and TRAIL-R2  (41, 78) 
 11q BIRC2 and BIRC3  (79, 80) 
 13 RB1 and DIS3  (41) 
 14q TRAF3  (79, 80) 
 16q CYLD and WWOX  (40, 41) 
 17p TP53  (41, 81) 
Epigenetic events 
Global hypomethylation (MGUS to multiple myeloma) and gene-specific hypermethylation (multiple myeloma to PC leukemia) Genome-wide methylation arrays (49)  

Abbreviation: MGUS, monoclonal gammopathy of undetermined significance.

The use of massively parallel sequencing has identified hundreds of mutations in multiple myeloma (42, 43); however, no consistently mutated gene has been identified that characterizes all occurrences of the disease. Nevertheless, these studies are in their infancy; the prognostic relevance of the multitude of exonic mutations that have been identified has yet to be determined. Although potentially useful, the challenge with interpreting sequencing data is defining which mutations are so-called “driver variants” that are important to disease pathogenesis rather than being “passenger variants” that are present by virtue of being carried forward by their association with other pathogenetically important genes. Driver mutations identified by this approach may be important therapeutic targets that could be used to design targeted treatment approaches.

Biomarker development

If we are to use tumor-acquired genetic variants as biomarkers, it is important to consider their specificity and sensitivity for identifying the clinical outcome of interest (e.g., PFS, OS, or side effects). There are now considerable data on the use of “FISH-detected” cytogenetic markers to predict clinical outcomes. What is clear from these data is that no single marker offers either good sensitivity or specificity for the prediction of either PFS or OS. If we are to alter the treatment following the use of a FISH-based cytogenetic approach, we will need to use a comprehensive panel of markers to define risk based on both the number and nature of the adverse markers detected. Interphase FISH (iFISH) combined with the ISS is useful, but general applicability and issues with sensitivity and specificity remain. Some of these challenges can be overcome by counting the number of abnormal lesions that are present and using the number present as a way of defining risk. This approach, combined with the use of a comprehensive panel of markers, including the adverse translocations t(4;14) and MAF together with the adverse copy number variables 1q+, 1p−, and 17p−, can provide very complete clinical data.

Global gene expression profiling (GEP) and prognostic signatures provide an alternative approach to FISH (44). Although GEP and prognostic signatures offer greater specificity in patients who are identified as having a poor prognosis, they lack biologic relevance and remain difficult to apply in a routine clinical setting. However, GEP does provide a massive amount of data and can define the groups identified in the translocation cyclin D classification (TC) as well as detect prognostic signatures. At this time, GEP cannot usefully identify the adverse groups defined by 1q+ and 17p−; these two subgroups with a poor prognosis still need to be detected by iFISH. The eventual clinical application and regular use of such biomarkers depends upon their clinical applicability, together with the availability of effective treatments for use in specific patient subgroups.

The technology used to detect a cytogenetic biomarker is also of critical importance. Historically, metaphase cytogenetic analysis was used, but this only gave results in 18% of patients and was therefore not widely adopted. Although FISH on CD138-selected PCs is applicable and gives results in nearly 100% of patients if adequate numbers of tumor cells are obtained, this method is slow and expensive, and up to now has lacked sensitivity and specificity. For predicting clinical outcomes, new genome-wide technologies are useful, yet their design and application are still in their infancy. Alternative strategies that may be more applicable for detecting copy number changes include multiplex ligation–dependent probe amplification (MLPA), which can detect the clinically relevant multiple myeloma–associated copy number variants at a fraction of the cost of FISH. MLPA can also deliver results in a more timely fashion, whereas GEP can define risk status based on poor prognosis signatures. An alternative expression-based approach is to design quantitative real-time PCR (qRT-PCR) assays that can identify TC groups and prognostically relevant genes (45). If these assays are combined with MLPA copy number variant assays, this approach can provide all of the relevant prognostic variants in a set of tests that are readily applicable.

MicroRNAs (miRNA), which are more stable than conventional RNA, can be detected in serum as well as in malignant PCs (46). In this context, most work on multiple myeloma biomarkers has focused on BMPCs; little work has been done on circulating PCs or serum DNA or RNA. These areas offer considerable opportunities for future development (3, 47). In addition, important epigenetic events—including global DNA hypomethylation and gene-specific DNA hypermethylation—are beginning to provide insights into the etiology of disease progression and could lead to the identification of clinically useful biomarkers (4, 48, 49).

The development of clinically relevant biomarkers is now being shaped by the regulatory framework for such testing. The Clinical Laboratory Improvement Amendments (CLIA) approach suggests that the development of biomarkers needs to be done in defined laboratories that work according to relevant standard operating procedures. This implies that all biomarkers need to be developed in the context of clinical trials in which data are collected according to the principles of good clinical practice, and the biomarkers are determined in a laboratory working as per CLIA standards. In the absence of such an approach, biomarkers cannot be adopted into clinical practice.

Risk-stratified treatment of multiple myeloma

The identification of patient-specific cytogenetic abnormalities and patients' gene mutation status may be particularly useful in helping to direct treatment in patients with multiple myeloma who possess a specific oncogenomic profile (5). It is now possible to define low-risk and high-risk disease subsets using GEP and FISH (50). GEP is useful in defining the molecular subtypes of multiple myeloma as well as being able to define signatures of high-risk disease (50–53).

On the basis of data analyses from platforms that are derived from completed studies, we are making progress toward improving the outcomes of patients with standard-risk disease. Nevertheless, we have made little progress toward treating or managing patients with high-risk disease. These observations argue in favor of moving toward the design of clinical trials that specifically develop treatments for high-risk disease. However, before we can design such studies, the biomarkers for robustly identifying these risk groups need to be validated in order for us to screen an adequate number of patients in the molecular subgroup of interest. This clinical trial approach will require us to fully embrace modern genomic technologies that are applicable in routine diagnostic facilities.

“Clinical response” as a biomarker.

Changes in BMPCs, paraprotein, and light chains form the basis of assessing patient response to therapy and could be used as an alternative treatment endpoint. The standard approach to using these data is laid out in the IMWG response criteria (54). The achievement of complete response (CR) is an important endpoint when assessing new therapies (Table 3); it seems likely that the deeper the level of response, the better the clinical outcome will be. Indeed, the achievement of a CR with no detectable disease is the essential prerequisite for a cure. Thus, the definition of stringent CR (sCR) and the criteria defining this state are of some importance. The IMWG defines sCR similarly to CR, but patients with multiple myeloma must also have a normal FLC ratio with no evidence of clonal BMPCs (assessed using immunohistochemistry or two- to four-color flow cytometry; ref. 54). A consensus is beginning to emerge on the most appropriate panel of antibodies and the strategy for detecting malignant PCs, with considerable enthusiasm being directed toward their use as a definitive clinical endpoint. Flow cytometry has considerable merit for monitoring residual disease because it offers sensitivity and does not require one to design patient-/clonospecific approaches; it can also be performed in most hematooncology diagnostic laboratories.

Table 3.

Partial list of response criteria from the European Group for Blood and Marrow Transplantation (EBMT) and IMWG

ParameterEBMT response criteria (82)IMWG uniform response criteriaa (54)
sCR 
  • Not defined

  
Patient meets all of the following criteria:
  • CR as defined

  • Normal FLC ratio

  • Absence of clonal PCs by immunohistochemistry or 2- to 4-color flow cytometry

  
CR Presence of all of the following:
  • Absence of M protein in serum and urine, measured by immunofixation, maintained for ≥6 weeks

  • <5% BMPCs

  • If absence of M protein is sustained for 6 weeks, it is not necessary to repeat the bone marrow examination except in patients with nonsecretory multiple myeloma

  • No increase in size or number of lytic bone lesions

  • Disappearance of soft tissue plasmacytomas

  
Patient meets all of the following criteria:
  • Negative immunofixation of serum and urine

  • Disappearance of any soft tissue plasmacytomas

  • ≤5% PCs in bone marrow

  
Immunophenotypic CR 
  • Not defined

  
Patient meets all of the following criteria:
  • sCR as defined

  • Absence of phenotypically aberrant PCs (clonal) in bone marrow with a minimum of 1 million total bone marrow cells analyzed by multiparametric flow cytometry (with >4 colors)

  
Molecular CR 
  • Not defined

  
Patient meets all of the following criteria:
  • CR as defined

  • Negative allele-specific oligonucleotide PCR (sensitivity 10−5)

  
VGPR 
  • Not defined

  
Patient meets one of the following criteria:
  • Serum and urine M-component detectable by immunofixation but not on electrophoresis

  • ≥90% reduction in serum M-component plus urine M-component <100 mg/24 hours

  
PR Patient meets all of the following criteria:
  • ≥50% reduction of serum M protein for ≥6 weeks

  • Reduction in 24-hour urinary light chain excretion either by ≥90% or to <200 mg for ≥6 weeks

  • Nonsecretory multiple myeloma only: ≥50% reduction of plasma cells in bone marrow aspirate for ≥6 weeks

  • ≥50% reduction in the size of soft tissue plasmacytomas

  • No increase in size or number of lytic bone lesions

  
Patient meets all of the following criteria:
  • ≥50% reduction of serum M protein and reduction in 24-hour urinary M protein by ≥90% or to <200 mg/24 h

  • If serum and urine M protein cannot be measured, a ≥50% decrease in the difference between involved and uninvolved FLC levels is required in place of the M protein criteria

  • If serum and urine M protein cannot be measured and serum free light assay also cannot be measured, ≥50% reduction in plasma cells is required in place of M protein, provided baseline BMPC percentage was ≥30%

  • In addition to the above listed criteria, if present at baseline, a ≥50% reduction in the size of soft tissue plasmacytomas is also required

  
ParameterEBMT response criteria (82)IMWG uniform response criteriaa (54)
sCR 
  • Not defined

  
Patient meets all of the following criteria:
  • CR as defined

  • Normal FLC ratio

  • Absence of clonal PCs by immunohistochemistry or 2- to 4-color flow cytometry

  
CR Presence of all of the following:
  • Absence of M protein in serum and urine, measured by immunofixation, maintained for ≥6 weeks

  • <5% BMPCs

  • If absence of M protein is sustained for 6 weeks, it is not necessary to repeat the bone marrow examination except in patients with nonsecretory multiple myeloma

  • No increase in size or number of lytic bone lesions

  • Disappearance of soft tissue plasmacytomas

  
Patient meets all of the following criteria:
  • Negative immunofixation of serum and urine

  • Disappearance of any soft tissue plasmacytomas

  • ≤5% PCs in bone marrow

  
Immunophenotypic CR 
  • Not defined

  
Patient meets all of the following criteria:
  • sCR as defined

  • Absence of phenotypically aberrant PCs (clonal) in bone marrow with a minimum of 1 million total bone marrow cells analyzed by multiparametric flow cytometry (with >4 colors)

  
Molecular CR 
  • Not defined

  
Patient meets all of the following criteria:
  • CR as defined

  • Negative allele-specific oligonucleotide PCR (sensitivity 10−5)

  
VGPR 
  • Not defined

  
Patient meets one of the following criteria:
  • Serum and urine M-component detectable by immunofixation but not on electrophoresis

  • ≥90% reduction in serum M-component plus urine M-component <100 mg/24 hours

  
PR Patient meets all of the following criteria:
  • ≥50% reduction of serum M protein for ≥6 weeks

  • Reduction in 24-hour urinary light chain excretion either by ≥90% or to <200 mg for ≥6 weeks

  • Nonsecretory multiple myeloma only: ≥50% reduction of plasma cells in bone marrow aspirate for ≥6 weeks

  • ≥50% reduction in the size of soft tissue plasmacytomas

  • No increase in size or number of lytic bone lesions

  
Patient meets all of the following criteria:
  • ≥50% reduction of serum M protein and reduction in 24-hour urinary M protein by ≥90% or to <200 mg/24 h

  • If serum and urine M protein cannot be measured, a ≥50% decrease in the difference between involved and uninvolved FLC levels is required in place of the M protein criteria

  • If serum and urine M protein cannot be measured and serum free light assay also cannot be measured, ≥50% reduction in plasma cells is required in place of M protein, provided baseline BMPC percentage was ≥30%

  • In addition to the above listed criteria, if present at baseline, a ≥50% reduction in the size of soft tissue plasmacytomas is also required

  

aComplete response (CR), stringent complete response (sCR), very good partial response (VGPR), and partial response (PR) require two consecutive assessments to be made at any time before beginning any new therapy. If radiographic studies were performed, these categories also require no known evidence of progressive or new bone lesions.

A recent survey that included 30 major medical institutions in the United States found that the application of multicolor flow cytometry for minimal residual disease (MRD) in multiple myeloma varies greatly (55). Indeed, the definition of abnormal PCs differed substantially among institutions, with some relying on CD19 and CD45 negativity with or without CD56 positivity to determine the extent of MRD, despite previous studies showing that normal PC subpopulations can be negative for CD19 and CD45 or CD56 positive (56, 57). More specific antigens such as CD27, CD81, and CD117 were used by less than half of the institutions. Even more importantly, there is considerable variation in the number of BMPCs analyzed (i.e., events) and the number of abnormal PCs that are needed to determine the presence of MRD, which affects the maximum possible sensitivity of the assay. In this survey, it was found that the maximum detection sensitivity ranged from 0.0005% to 0.02%, a 100-fold difference in sensitivity. Such data copy the establishment of standardized approaches and external quality control programs. It is important to contrast this with multiparameter flow cytometry with the application of clonospecific PCR for the same purpose. In most settings, the clonospecific PCR offers greater sensitivity, but this technique has been hampered by the necessity to develop clonospecific probes, which has made its general application cumbersome and expensive. This situation is now changing with the advent of massively parallel sequencing approaches, which use probes that can be used to tile entire immunoglobulin heavy chain gene locus (IgH) regions (43). This approach is sensitive, high throughput, and universally applicable.

Predictive biomarkers.

In recent years, it has become evident that patients with the t(4;14) have more favorable outcomes when they are treated with proteasome inhibitors. A group at the Mayo Clinic recently discussed the concept of “risk-adapted therapy” (5), whereby patients with high-risk disease [i.e., patients with the t(14;16), t(14;20), and/or del17p] may be better candidates for triplet combination therapy (e.g., bortezomib–lenalidomide–dexamethasone) compared with patients with intermediate-risk or standard-risk disease. Although triplet combinations may have a less favorable side-effect profile relative to single-agent therapies, triplet combination regimens are more appropriate for patients with high-risk disease because median OS in that cohort is only 3 years compared with 4 to 5 years and 8 to 10 years for patients with intermediate-risk and standard-risk disease, respectively. In the absence of using cytogenetic markers to risk-stratify transplant-eligible and transplant-ineligible patients, it would be difficult to determine which treatment regimen is most appropriate for patients with multiple myeloma.

Nevertheless, the t(4;14) cannot be considered to be a true prognostic biomarker because many patients who lack this translocation also respond and have good clinical outcomes following proteasome inhibitor therapy. Moving forward, information from genome-wide sequencing studies is leading to the identification of frequent pathogenetically important mutations in multiple myeloma, including those involved in the extracellular signal-regulated kinase (ERK) signal transduction pathway. These biomarkers include NRAS, KRAS, and BRAF (4). In recent years, BRAF-V600E has emerged as one of the most promising ERK mutations that can be targeted, validating the concept of targeted treatment. For example, a patient with multiple myeloma with extramedullary disease and the BRAF-V600E mutation responded well to low doses of vemurafenib, a mutation-specific BRAF inhibitor (58), making it a true predictive biomarker in the absence of which a response was not observed.

One potential issue with use of targeted therapy is the presence of ICH relating to subclonal variability. ICH is the essential substrate for clonal evolution according to the principles of Darwinian evolution (4). In recent years, three patterns of clonal evolution have been identified: relapse can be genetically stable; it can linearly evolve with several new genetic variants at relapse; or it can come from a clone that was not present at the time of diagnosis (59). Paired sample analysis of 28 patients with multiple myeloma revealed that patients with standard-risk cytogenetics typically have PC clones that are genetically stable (59). In contrast, patients with high-risk cytogenetics have PC clones that undergo many more genetic modifications over time.

The presence of ICH has important implications for the development of biomarkers. Although it was once acceptable to determine whether a molecular target was present, it is now essential to obtain an idea of the size of the subclone carrying the target. The size of the clone has important therapeutic implications for targeted treatment because completely eradicating a clone present only 5% of the time would have little importance clinically, whereas in a clone present 90% of the time, there would be a significant clinical response.

The presence of ICH also has an effect on the assessment and treatment of relapse. Traditionally, clinical data were used to define prognosis, response to therapy, duration of prior response, rate of relapse, the presence of extramedullary or blastic disease, performance status, and the toxicity of prior treatment. In the era of molecular medicine, cytogenetics, focal lesions, tidal clone status (i.e., same clone, new mutant, or prior clone), GEP signature, and methylation status are beginning to provide clinicians with more information about the clinical behavior of the disease at relapse.

Biomarkers in monoclonal gammopathy of undetermined significance and smoldering multiple myeloma

In the 1970s, Kyle and Greipp began to use the phrases monoclonal gammopathy of undetermined significance (MGUS) and smoldering multiple myeloma (SMM) to describe premalignant PC disorders that are not associated with end-organ damage or treatment (60, 61). Currently, asymptomatic multiple myeloma is an active area of biomarker research in which the challenge is to predict those patients who are at high risk of disease progression for whom treatment intervention is essential to prevent the emergence of significant end-organ damage. The use of many of the molecular markers described above is difficult because the molecular features of multiple myeloma that require treatment are often present in patients with asymptomatic disease (62, 63), meaning that alternative approaches to predict risk status are required.

In 2003, the IMWG used laboratory and clinical markers (e.g., serum and urine M protein, clonal BMPCs, and sFLC) to define patients with asymptomatic multiple myeloma in which treatment was not indicated (64). The sFLC ratio is one of the most promising biomarkers in asymptomatic multiple myeloma; it has been used as a prognostic indicator in patients with MGUS (65), SMM (66–68), and newly diagnosed multiple myeloma (69), but it is not without controversy. The Spanish Programa Español de Tratamientos en Hematología (PETHEMA) and Mayo Clinic groups both used sFLC ratios, circulating PCs, and PC proliferation rates to create two models that classify asymptomatic patients as being at low, intermediate, or high risk of disease progression (66, 70). A group at the NCI used the Spanish PETHEMA model (70) and the Mayo Clinic model (66) to categorize 77 patients with SMM as being at low, intermediate, or high risk of developing active multiple myeloma (71). In the NCI's study, concordance between the PETHEMA and Mayo Clinic models was low (28.6%); there was also significant discordance between the ways in which each model classified patients' risk status. Thus, although characterization of PC immunophenotype and measuring sFLC ratios both have prognostic potential, they lack specificity and do not have enough positive predictive value to be useful in determining when treatment should be initiated in patients with asymptomatic disease.

Imaging tools for early myeloma

Given that nearly all patients with symptomatic multiple myeloma develop osteolytic lesions, clinicians use bone marrow aspirates and trephine samples to monitor bone morphology throughout the course of disease progression. Although these tools are useful, there can be variability in the distribution of BMPCs throughout the body, increasing the likelihood of heterogeneous sample recovery and sampling error, particularly in patients with asymptomatic disease. Given the importance of understanding the underlying biology of bones in multiple myeloma, clinicians use X-rays to complete skeletal surveys, a technique that is currently the gold standard for evaluating severity of bone disease. Despite the utility of skeletal surveys, MRI, computed tomography (CT), and positron emission tomography (PET)/CT scanning are becoming increasingly important tools for assessment of disease severity. MRI is a useful noninvasive technique for imaging the spine, soft tissue, and infiltration of BMPCs, and is particularly useful for patients with SMM because it can detect diffuse lesions, the presence of which is an adverse prognostic factor for PFS (72). In addition, detection of seven or more focal lesions with MRI is considered to be an adverse prognostic factor in patients with multiple myeloma (73). If MRI is unavailable, CT may be helpful for assessing the extent of damage to the spine or soft tissues. Nevertheless, its use should be limited because CT can expose patients to nearly three times more radiation than conventional MRI. Finally, fusion of CT and fluorodeoxyglucose (FDG) PET images can be very useful for the evaluation of patients with symptomatic disease and has the potential to be useful in patients with MGUS or SMM (74). In the future, functional imaging techniques that are used in combination with biomarker data are likely to contribute to the management of patients with “early” multiple myeloma and the development of novel MRD assays.

Cytotoxic chemotherapy, immunomodulatory agents, and proteasome inhibitors have been highly effective and universally applicable in multiple myeloma. Going forward, the next generation of targeted treatments will depend not only on the presence of a target, but also on their ability to modulate their target. This will be especially important during the early phases of clinical development of targeted treatments, in which target modulation should be considered to be an important endpoint. In multiple myeloma, it is, perhaps, not optimal to use circulating lymphocytes as targets to assess target modulation because of differences in cellular biology and tissue penetration. Consequently, access to and characterization of BMPCs is important and can be achieved using immunohistochemistry on bone marrow trephines or flow cytometry on aspirate samples. Examples include the assessment of antiapoptotic proteins to predict a patient's response to agents that target these pathways, the demonstration of downregulation of pERK, or evidence of changes in histone methylation states following evaluation of MMSET inhibitors.

Over the past 40 years, biomarkers such as M protein and serum β2M have contributed innumerable insights into our present understanding of multiple myeloma. Today, many cytogenetic markers are beginning to provide information about the severity of the disease; these markers are also beginning to inform clinicians about which anti-multiple myeloma treatment regimen is most appropriate for a particular patient. It is clear that rapid technological advances are changing the way biomarkers are perceived as well as their clinical relevance (Fig. 1). In the near future, diagnostic tests based on massively parallel sequencing approaches will enable detection of recurrent molecular abnormalities as well as actionable mutations that are rarely observed but which could use treatments that are used in other disease settings. Massively parallel sequencing approaches will also enable clinicians to monitor disease longitudinally, which may provide opportunities for early intervention before clonal PC expansion and disease complexity render existing treatments ineffective. Finally, combining functional imaging techniques with assays that use biomarkers has the potential to change clinical practice in patients with early myeloma and patients with symptomatic disease.

Figure 1.

Technological advances in detecting biomarkers in multiple myeloma.

Figure 1.

Technological advances in detecting biomarkers in multiple myeloma.

Close modal

No potential conflicts of interest were disclosed.

Conception and design: O. Landgren, G.J. Morgan

Development of methodology: O. Landgren, G.J. Morgan

Acquisition of data (provided animals, acquired and managed patients, provided facilities, etc.): O. Landgren, G.J. Morgan

Analysis and interpretation of data (e.g., statistical analysis, biostatistics, computational analysis): O. Landgren, G.J. Morgan

Writing, review, and/or revision of the manuscript: O. Landgren, G.J. Morgan

Administrative, technical, or material support (i.e., reporting or organizing data, constructing databases): G.J. Morgan

Study supervision: O. Landgren, G.J. Morgan

The authors thank A. Peter Morello and Thomas Renau (Onyx Pharmaceuticals, Inc., South San Francisco, CA) for providing medical writing and editorial assistance, which was funded by Onyx Pharmaceuticals, Inc.

This study was supported by Celgene, Janssen, Novartis, Onyx, and Millennium (honoraria; to G.J. Morgan).

1.
Kumar
SK
,
Rajkumar
SV
,
Dispenzieri
A
,
Lacy
MQ
,
Hayman
SR
,
Buadi
FK
, et al
Improved survival in multiple myeloma and the impact of novel therapies
.
Blood
2008
;
111
:
2516
20
.
2.
Pessoa-Magalhaes
RJ
,
Vidriales
MB
,
Paiva
B
,
Gimenez
CF
,
Garcia-Sanz
R
,
Mateos
MV
, et al
Analysis of the immune system of multiple myeloma patients achieving long-term disease control, by multidimensional flow cytometry
.
Haematologica
2012
;
98
:
79
86
.
3.
Egan
JB
,
Shi
CX
,
Tembe
W
,
Christoforides
A
,
Kurdoglu
A
,
Sinari
S
, et al
Whole-genome sequencing of multiple myeloma from diagnosis to plasma cell leukemia reveals genomic initiating events, evolution, and clonal tides
.
Blood
2012
;
120
:
1060
6
.
4.
Morgan
GJ
,
Walker
BA
,
Davies
FE
. 
The genetic architecture of multiple myeloma
.
Nat Rev Cancer
2012
;
12
:
335
48
.
5.
Mikhael
JR
,
Dingli
D
,
Roy
V
,
Reeder
CB
,
Buadi
FK
,
Hayman
SR
, et al
Management of newly diagnosed symptomatic multiple myeloma: updated Mayo stratification of myeloma and risk-adapted therapy (mSMART) consensus guidelines 2013
.
Mayo Clin Proc
2013
;
88
:
360
76
.
6.
National Cancer Institute (NCI)
[database on the Internet]
.
Bethesda, MD
:
NCI
[cited 2013 Mar 25]. Available from
: http://www.cancer.gov/dictionary?cdrid=45618
7.
Carbone
PP
,
Kellerhouse
LE
,
Gehan
EA
. 
Plasmacytic myeloma: a study of the relationship of survival to various clinical manifestations and anomalous protein type in 112 patients
.
Am J Med
1967
;
42
:
937
48
.
8.
Costa
G
,
Engle
RL
,
Schilling
A
,
Carbone
P
,
Kochwa
S
,
Nachman
RL
, et al
Melphalan and prednisone: an effective combination for the treatment of multiple myeloma
.
Am J Med
1973
;
54
:
589
99
.
9.
Durie
BG
,
Salmon
SE
. 
A clinical staging system for multiple myeloma. Correlation of measured myeloma cell mass with presenting clinical features, response to treatment, and survival
.
Cancer
1975
;
36
:
842
54
.
10.
Bataille
R
,
Durie
BG
,
Grenier
J
. 
Serum β2-microglobulin and survival duration in multiple myeloma: a simple reliable marker for staging
.
Br J Haematol
1983
;
55
:
439
47
.
11.
Durie
BG
,
Stock-Novack
D
,
Salmon
SE
,
Finley
P
,
Beckord
J
,
Crowley
J
, et al
Prognostic value of pretreatment serum β2-microglobulin in myeloma: a Southwest Oncology Group Study
.
Blood
1990
;
75
:
823
30
.
12.
Bataille
R
,
Durie
BG
,
Grenier
J
,
Sany
J
. 
Prognostic factors and staging in multiple myeloma: a reappraisal
.
J Clin Oncol
1986
;
4
:
80
7
.
13.
Bataille
R
,
Boccadoro
M
,
Klein
B
,
Durie
B
,
Pileri
A
. 
C-reactive protein and β2-microglobulin produce a simple and powerful myeloma staging system
.
Blood
1992
;
80
:
733
7
.
14.
Greipp
PR
,
Lust
JA
,
O'Fallon
WM
,
Katzmann
JA
,
Witzig
TE
,
Kyle
RA
. 
Plasma cell labeling index and β2-microglobulin predict survival independent of thymidine kinase and C-reactive protein in multiple myeloma
.
Blood
1993
;
81
:
3382
7
.
15.
San Miguel
JF
,
Garcia-Sanz
R
,
Gonzalez
M
,
Moro
MJ
,
Hernandez
JM
,
Ortega
F
, et al
A new staging system for multiple myeloma based on the number of S-phase plasma cells
.
Blood
1995
;
85
:
448
55
.
16.
Greipp
PR
,
San Miguel
J
,
Durie
BG
,
Crowley
JJ
,
Barlogie
B
,
Blade
J
, et al
International staging system for multiple myeloma
.
J Clin Oncol
2005
;
23
:
3412
20
.
17.
Boyd
KD
,
Ross
FM
,
Chiecchio
L
,
Dagrada
GP
,
Konn
ZJ
,
Tapper
WJ
, et al
A novel prognostic model in myeloma based on co-segregating adverse FISH lesions and the ISS: analysis of patients treated in the MRC myeloma IX trial
.
Leukemia
2012
;
26
:
349
55
.
18.
Avet-Loiseau
H
,
Durie
BG
,
Cavo
M
,
Attal
M
,
Gutierrez
N
,
Haessler
J
, et al
Combining fluorescent in situ hybridization data with ISS staging improves risk assessment in myeloma: an International Myeloma Working Group collaborative project
.
Leukemia
2013
;
27
:
711
7
.
19.
Fonseca
R
,
Bergsagel
PL
,
Drach
J
,
Shaughnessy
J
,
Gutierrez
N
,
Stewart
AK
, et al
International Myeloma Working Group molecular classification of multiple myeloma: spotlight review
.
Leukemia
2009
;
23
:
2210
21
.
20.
Ludwig
H
,
Milosavljevic
D
,
Zojer
N
,
Faint
JM
,
Bradwell
AR
,
Hubl
W
, et al
Immunoglobulin heavy/light chain ratios improve paraprotein detection and monitoring, identify residual disease and correlate with survival in multiple myeloma patients
.
Leukemia
2012
;
27
:
213
9
.
21.
Chesi
M
,
Bergsagel
PL
,
Brents
LA
,
Smith
CM
,
Gerhard
DS
,
Kuehl
WM
. 
Dysregulation of cyclin D1 by translocation into an IgH gamma switch region in two multiple myeloma cell lines
.
Blood
1996
;
88
:
674
81
.
22.
Bergsagel
PL
,
Kuehl
WM
. 
Chromosome translocations in multiple myeloma
.
Oncogene
2001
;
20
:
5611
22
.
23.
Shaughnessy
J
 Jr
,
Gabrea
A
,
Qi
Y
,
Brents
L
,
Zhan
F
,
Tian
E
, et al
Cyclin D3 at 6p21 is dysregulated by recurrent chromosomal translocations to immunoglobulin loci in multiple myeloma
.
Blood
2001
;
98
:
217
23
.
24.
Hurt
EM
,
Wiestner
A
,
Rosenwald
A
,
Shaffer
AL
,
Campo
E
,
Grogan
T
, et al
Overexpression of c-maf is a frequent oncogenic event in multiple myeloma that promotes proliferation and pathological interactions with bone marrow stroma
.
Cancer Cell
2004
;
5
:
191
9
.
25.
Bergsagel
PL
,
Kuehl
WM
,
Zhan
F
,
Sawyer
J
,
Barlogie
B
,
Shaughnessy
J
 Jr
. 
Cyclin D dysregulation: an early and unifying pathogenic event in multiple myeloma
.
Blood
2005
;
106
:
296
303
.
26.
Ross
FM
,
Chiecchio
L
,
Dagrada
G
,
Protheroe
RK
,
Stockley
DM
,
Harrison
CJ
, et al
The t(14;20) is a poor prognostic factor in myeloma but is associated with long-term stable disease in monoclonal gammopathies of undetermined significance
.
Haematologica
2010
;
95
:
1221
5
.
27.
Weinhold
N
,
Johnson
DC
,
Chubb
D
,
Chen
B
,
Forsti
A
,
Hosking
FJ
, et al
The CCND1 c.870G>A polymorphism is a risk factor for t(11;14)(q13;q32) multiple myeloma
.
Nat Genet
2013
;
45
:
522
5
.
28.
Fonseca
R
,
Blood
E
,
Rue
M
,
Harrington
D
,
Oken
MM
,
Kyle
RA
, et al
Clinical and biologic implications of recurrent genomic aberrations in myeloma
.
Blood
2003
;
101
:
4569
75
.
29.
Avet-Loiseau
H
,
Attal
M
,
Moreau
P
,
Charbonnel
C
,
Garban
F
,
Hulin
C
, et al
Genetic abnormalities and survival in multiple myeloma: the experience of the Intergroupe Francophone du Myelome
.
Blood
2007
;
109
:
3489
95
.
30.
Oswald
F
,
Lovec
H
,
Moroy
T
,
Lipp
M
. 
E2F-dependent regulation of human MYC: trans-activation by cyclins D1 and A overrides tumour suppressor protein functions
.
Oncogene
1994
;
9
:
2029
36
.
31.
Hoyer
JD
,
Hanson
CA
,
Fonseca
R
,
Greipp
PR
,
Dewald
GW
,
Kurtin
PJ
. 
The (11;14)(q13;q32) translocation in multiple myeloma. A morphologic and immunohistochemical study
.
Am J Clin Pathol
2000
;
113
:
831
7
.
32.
Garand
R
,
Avet-Loiseau
H
,
Accard
F
,
Moreau
P
,
Harousseau
JL
,
Bataille
R
. 
t(11;14) and t(4;14) translocations correlated with mature lymphoplasmacytoid and immature morphology, respectively, in multiple myeloma
.
Leukemia
2003
;
17
:
2032
5
.
33.
Chng
WJ
,
Huang
GF
,
Chung
TH
,
Ng
SB
,
Gonzalez-Paz
N
,
Troska-Price
T
, et al
Clinical and biological implications of MYC activation: a common difference between MGUS and newly diagnosed multiple myeloma
.
Leukemia
2011
;
25
:
1026
35
.
34.
Broderick
P
,
Chubb
D
,
Johnson
DC
,
Weinhold
N
,
Forsti
A
,
Lloyd
A
, et al
Common variation at 3p22.1 and 7p15.3 influences multiple myeloma risk
.
Nat Genet
2012
;
44
:
58
61
.
35.
Martino
A
,
Campa
D
,
Jamroziak
K
,
Reis
RM
,
Sainz
J
,
Buda
G
, et al
Impact of polymorphic variation at 7p15.3, 3p22.1 and 2p23.3 loci on risk of multiple myeloma
.
Br J Haematol
2012
;
158
:
805
9
.
36.
Rajkumar
SV
,
Gupta
V
,
Fonseca
R
,
Dispenzieri
A
,
Gonsalves
WI
,
Larson
D
, et al
Impact of primary molecular cytogenetic abnormalities and risk of progression in smoldering multiple myeloma
.
Leukemia
2013
;
27
:
1738
44
.
37.
Onodera
N
,
McCabe
NR
,
Rubin
CM
. 
Formation of a hyperdiploid karyotype in childhood acute lymphoblastic leukemia
.
Blood
1992
;
80
:
203
8
.
38.
Ross
RW
,
Oh
WK
,
Xie
W
,
Pomerantz
M
,
Nakabayashi
M
,
Sartor
O
, et al
Inherited variation in the androgen pathway is associated with the efficacy of androgen-deprivation therapy in men with prostate cancer
.
J Clin Oncol
2008
;
26
:
842
7
.
39.
Avet-Loiseau
H
,
Li
C
,
Magrangeas
F
,
Gouraud
W
,
Charbonnel
C
,
Harousseau
JL
, et al
Prognostic significance of copy-number alterations in multiple myeloma
.
J Clin Oncol
2009
;
27
:
4585
90
.
40.
Jenner
MW
,
Leone
PE
,
Walker
BA
,
Ross
FM
,
Johnson
DC
,
Gonzalez
D
, et al
Gene mapping and expression analysis of 16q loss of heterozygosity identifies WWOX and CYLD as being important in determining clinical outcome in multiple myeloma
.
Blood
2007
;
110
:
3291
300
.
41.
Walker
BA
,
Leone
PE
,
Chiecchio
L
,
Dickens
NJ
,
Jenner
MW
,
Boyd
KD
, et al
A compendium of myeloma-associated chromosomal copy number abnormalities and their prognostic value
.
Blood
2010
;
116
:
e56
65
.
42.
Chapman
MA
,
Lawrence
MS
,
Keats
JJ
,
Cibulskis
K
,
Sougnez
C
,
Schinzel
AC
, et al
Initial genome sequencing and analysis of multiple myeloma
.
Nature
2011
;
471
:
467
72
.
43.
Walker
BA
,
Wardell
CP
,
Johnson
DC
,
Kaiser
MF
,
Begum
DB
,
Dahir
NB
, et al
Characterization of IGH locus breakpoints in multiple myeloma indicates a subset of translocations appear to occur in pregerminal center B cells
.
Blood
2013
;
121
:
3413
9
.
44.
Kuiper
R
,
Broyl
A
,
de Knegt
Y
,
van Vliet
MH
,
van Beers
EH
,
van der Holt
B
, et al
A gene expression signature for high-risk multiple myeloma
.
Leukemia
2012
;
26
:
2406
13
.
45.
Kaiser
MF
,
Walker
BA
,
Hockley
SL
,
Begum
DB
,
Wardell
CP
,
Gonzalez
D
, et al
A TC classification-based predictor for multiple myeloma using multiplexed real-time quantitative PCR
.
Leukemia
2013
;
27
:
1754
7
.
46.
Jones
CI
,
Zabolotskaya
MV
,
King
AJ
,
Stewart
HJ
,
Horne
GA
,
Chevassut
TJ
, et al
Identification of circulating microRNAs as diagnostic biomarkers for use in multiple myeloma
.
Br J Cancer
2012
;
107
:
1987
96
.
47.
Calvo
KR
,
Landgren
O
,
Roccaro
AM
,
Ghobrial
IM
. 
Role of microRNAs from monoclonal gammopathy of undetermined significance to multiple myeloma
.
Semin Hematol
2011
;
48
:
39
45
.
48.
Cedar
H
,
Bergman
Y
. 
Linking DNA methylation and histone modification: patterns and paradigms
.
Nat Rev Genet
2009
;
10
:
295
304
.
49.
Walker
BA
,
Wardell
CP
,
Chiecchio
L
,
Smith
EM
,
Boyd
KD
,
Neri
A
, et al
Aberrant global methylation patterns affect the molecular pathogenesis and prognosis of multiple myeloma
.
Blood
2011
;
117
:
553
62
.
50.
Sawyer
JR
. 
The prognostic significance of cytogenetics and molecular profiling in multiple myeloma
.
Cancer Genet
2011
;
204
:
3
12
.
51.
Zhan
F
,
Tian
E
,
Bumm
K
,
Smith
R
,
Barlogie
B
,
Shaughnessy
J
 Jr
. 
Gene expression profiling of human plasma cell differentiation and classification of multiple myeloma based on similarities to distinct stages of late-stage B-cell development
.
Blood
2003
;
101
:
1128
40
.
52.
Broyl
A
,
Hose
D
,
Lokhorst
H
,
de Knegt
Y
,
Peeters
J
,
Jauch
A
, et al
Gene expression profiling for molecular classification of multiple myeloma in newly diagnosed patients
.
Blood
2010
;
116
:
2543
53
.
53.
Shaughnessy
JD
 Jr
,
Qu
P
,
Usmani
S
,
Heuck
CJ
,
Zhang
Q
,
Zhou
Y
, et al
Pharmacogenomics of bortezomib test-dosing identifies hyperexpression of proteasome genes, especially PSMD4, as novel high-risk feature in myeloma treated with Total Therapy 3
.
Blood
2011
;
118
:
3512
24
.
54.
Durie
BG
,
Harousseau
JL
,
Miguel
JS
,
Blade
J
,
Barlogie
B
,
Anderson
K
, et al
International uniform response criteria for multiple myeloma
.
Leukemia
2006
;
20
:
1467
73
.
55.
Flanders
A
,
Stetler-Stevenson
M
,
Landgren
O
. 
Minimal residual disease testing in multiple myeloma by flow cytometry: major heterogeneity
.
Blood
2013
;
122
:
1088
9
.
56.
Cannizzo
E
,
Bellio
E
,
Sohani
AR
,
Hasserjian
RP
,
Ferry
JA
,
Dorn
ME
, et al
Multiparameter immunophenotyping by flow cytometry in multiple myeloma: the diagnostic utility of defining ranges of normal antigenic expression in comparison to histology
.
Cytometry B Clin Cytom
2010
;
78
:
231
8
.
57.
Liu
D
,
Lin
P
,
Hu
Y
,
Zhou
Y
,
Tang
G
,
Powers
L
, et al
Immunophenotypic heterogeneity of normal plasma cells: comparison with minimal residual plasma cell myeloma
.
J Clin Pathol
2012
;
65
:
823
9
.
58.
Andrulis
M
,
Lehners
N
,
Capper
D
,
Penzel
R
,
Heining
C
,
Huellein
J
, et al
Targeting the BRAF V600E mutation in multiple myeloma
.
Cancer Discov
2013
;
3
:
862
9
.
59.
Keats
JJ
,
Chesi
M
,
Egan
JB
,
Garbitt
VM
,
Palmer
SE
,
Braggio
E
, et al
Clonal competition with alternating dominance in multiple myeloma
.
Blood
2012
;
120
:
1067
76
.
60.
Kyle
RA
. 
Monoclonal gammopathy of undetermined significance. Natural history in 241 cases
.
Am J Med
1978
;
64
:
814
26
.
61.
Kyle
RA
,
Greipp
PR
. 
Smoldering multiple myeloma
.
N Engl J Med
1980
;
302
:
1347
9
.
62.
Zandecki
M
,
Lai
JL
,
Genevieve
F
,
Bernardi
F
,
Volle-Remy
H
,
Blanchet
O
, et al
Several cytogenetic subclones may be identified within plasma cells from patients with monoclonal gammopathy of undetermined significance, both at diagnosis and during the indolent course of this condition
.
Blood
1997
;
90
:
3682
90
.
63.
Avet-Loiseau
H
,
Facon
T
,
Daviet
A
,
Godon
C
,
Rapp
MJ
,
Harousseau
JL
, et al
14q32 translocations and monosomy 13 observed in monoclonal gammopathy of undetermined significance delineate a multistep process for the oncogenesis of multiple myeloma. Intergroupe Francophone du Myelome
.
Cancer Res
1999
;
59
:
4546
50
.
64.
International Myeloma Working Group
. 
Criteria for the classification of monoclonal gammopathies, multiple myeloma and related disorders: a report of the International Myeloma Working Group
.
Br J Haematol
2003
;
121
:
749
57
.
65.
Rajkumar
SV
,
Kyle
RA
,
Therneau
TM
,
Melton
LJ
 III
,
Bradwell
AR
,
Clark
RJ
, 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
.
66.
Dispenzieri
A
,
Kyle
RA
,
Katzmann
JA
,
Therneau
TM
,
Larson
D
,
Benson
J
, et al
Immunoglobulin free light chain ratio is an independent risk factor for progression of smoldering (asymptomatic) multiple myeloma
.
Blood
2008
;
111
:
785
9
.
67.
Larsen
JT
,
Kumar
SK
,
Dispenzieri
A
,
Kyle
RA
,
Katzmann
JA
,
Rajkumar
SV
. 
Serum free light chain ratio as a biomarker for high-risk smoldering multiple myeloma
.
Leukemia
2012
;
27
:
941
6
.
68.
Iwama
K
,
Chihara
D
,
Tsuda
K
,
Ugai
T
,
Sugihara
H
,
Nishida
Y
, et al
Normalization of free light chain kappa/lambda ratio is a robust prognostic indicator of favorable outcome in patients with multiple myeloma
.
Eur J Haematol
2013
;
90
:
134
41
.
69.
Snozek
CL
,
Katzmann
JA
,
Kyle
RA
,
Dispenzieri
A
,
Larson
DR
,
Therneau
TM
, et al
Prognostic value of the serum free light chain ratio in newly diagnosed myeloma: proposed incorporation into the international staging system
.
Leukemia
2008
;
22
:
1933
7
.
70.
Perez-Persona
E
,
Vidriales
MB
,
Mateo
G
,
Garcia-Sanz
R
,
Mateos
MV
,
de Coca
AG
, et al
New criteria to identify risk of progression in monoclonal gammopathy of uncertain significance and smoldering multiple myeloma based on multiparameter flow cytometry analysis of bone marrow plasma cells
.
Blood
2007
;
110
:
2586
92
.
71.
Cherry
BM
,
Korde
N
,
Kwok
M
,
Manasanch
EE
,
Bhutani
M
,
Mulquin
M
, et al
Modeling progression risk for smoldering multiple myeloma: results from a prospective clinical study
.
Leuk Lymphoma
2013
;
54
:
2215
8
.
72.
Hillengass
J
,
Fechtner
K
,
Weber
MA
,
Bauerle
T
,
Ayyaz
S
,
Heiss
C
, et al
Prognostic significance of focal lesions in whole-body magnetic resonance imaging in patients with asymptomatic multiple myeloma
.
J Clin Oncol
2010
;
28
:
1606
10
.
73.
Walker
R
,
Barlogie
B
,
Haessler
J
,
Tricot
G
,
Anaissie
E
,
Shaughnessy
JD
 Jr
, et al
Magnetic resonance imaging in multiple myeloma: diagnostic and clinical implications
.
J Clin Oncol
2007
;
25
:
1121
8
.
74.
Landgren
O
,
Waxman
AJ
. 
Multiple myeloma precursor disease
.
JAMA
2010
;
304
:
2397
404
.
75.
Avet-Loiseau
H
,
Gerson
F
,
Magrangeas
F
,
Minvielle
S
,
Harousseau
JL
,
Bataille
R
. 
Rearrangements of the c-myc oncogene are present in 15% of primary human multiple myeloma tumors
.
Blood
2001
;
98
:
3082
6
.
76.
Cigudosa
JC
,
Rao
PH
,
Calasanz
MJ
,
Odero
MD
,
Michaeli
J
,
Jhanwar
SC
, et al
Characterization of nonrandom chromosomal gains and losses in multiple myeloma by comparative genomic hybridization
.
Blood
1998
;
91
:
3007
10
.
77.
Leone
PE
,
Walker
BA
,
Jenner
MW
,
Chiecchio
L
,
Dagrada
G
,
Protheroe
RK
, et al
Deletions of CDKN2C in multiple myeloma: biological and clinical implications
.
Clin Cancer Res
2008
;
14
:
6033
41
.
78.
Gmidene
A
,
Saad
A
,
Avet-Loiseau
H
. 
8p21.3 deletion suggesting a probable role of TRAIL-R1 and TRAIL-R2 as candidate tumor suppressor genes in the pathogenesis of multiple myeloma
.
Med Oncol
2013
;
30
:
489
.
79.
Annunziata
CM
,
Davis
RE
,
Demchenko
Y
,
Bellamy
W
,
Gabrea
A
,
Zhan
F
, et al
Frequent engagement of the classical and alternative NF-κB pathways by diverse genetic abnormalities in multiple myeloma
.
Cancer Cell
2007
;
12
:
115
30
.
80.
Keats
JJ
,
Fonseca
R
,
Chesi
M
,
Schop
R
,
Baker
A
,
Chng
WJ
, et al
Promiscuous mutations activate the noncanonical NF-κB pathway in multiple myeloma
.
Cancer Cell
2007
;
12
:
131
44
.
81.
Boyd
KD
,
Ross
FM
,
Tapper
WJ
,
Chiecchio
L
,
Dagrada
G
,
Konn
ZJ
, et al
The clinical impact and molecular biology of del(17p) in multiple myeloma treated with conventional or thalidomide-based therapy
.
Genes Chromosomes Cancer
2011
;
50
:
765
74
.
82.
Blade
J
,
Samson
D
,
Reece
D
,
Apperley
J
,
Bjorkstrand
B
,
Gahrton
G
, et al
Criteria for evaluating disease response and progression in patients with multiple myeloma treated by high-dose therapy and haemopoietic stem cell transplantation. Myeloma Subcommittee of the EBMT. European Group for Blood and Marrow Transplant
.
Br J Haematol
1998
;
102
:
1115
23
.