The term monoclonal gammopathy of undetermined significance (MGUS) was coined in 1978. The recent advances in our knowledge about MGUS and smoldering multiple myeloma (SMM) have helped us better understand the pathogenesis of myeloma. It seems that myeloma evolves from a precursor state in almost all cases. We do not completely understand the multistep process from the precursor state to myeloma, but studies like whole genome sequencing continue to improve our understanding of this process. The process of transformation may not be linear acquisition of changes, but rather a branched heterogeneous process. Clinical features that are prognostic of rapid transformation have been identified, but no specific molecular markers have been identified. Even with recent advances, multiple myeloma remains an incurable disease in the vast majority, and intervening at the precursor state provides a unique opportunity to alter the natural history of the disease. A limitation is that a vast majority of patients with precursor disease, especially low-risk MGUS, will never progress to myeloma in their lifetime, and treating these patients is not only unnecessary but may be potentially harmful. The challenge is to identify a subset of patients with the precursor state that would definitely progress to myeloma and in whom interventions will have a meaningful impact. As our understanding of the molecular and genetic processes improves, these studies will guide the selection of high-risk patients more appropriately and ultimately direct a tailored management strategy to either delay progression to symptomatic myeloma or even “cure” a person at this premalignant stage. Clin Cancer Res; 19(5); 985–94. ©2012 AACR.

Multiple myeloma is a plasma cell neoplasm characterized by multifocal proliferation of clonal, long-lived plasma cells associated with an overproduction of monoclonal gammaglobulin (1). In 1961, Jan Walderström decribed “essential benign hypergammaglobulinemia” as an asymptomatic condition wherein the monoclonal gammopathy is not associated with any symptoms (2). The term monoclonal gammopathy of undertermined significance (MGUS) was coined in 1978 (3) and smoldering multiple myeloma (SMM) in 1980 (4). It has always been recognized that some cases of MGUS progressed to symptomatic multiple myeloma, but recent studies have shown that multiple myeloma is consistently preceded by MGUS (5, 6). This has given rise to the concept of a myeloma precursor disease and raised questions about the biologic events leading to progression of these precursor states to symptomatic myeloma. We will review the current understanding of the biology of these precursor states and also discuss trials looking at interventions.

In 2010, the International Myeloma Working Group (IMWG) defined MGUS by the presence of serum M-protein (< 3g/dL), clonal plasma cell population in the bone marrow (< 10%), and the absence of end-organ damage such as hypercalcemia (serum calcium ≥ 11.5 mg/dL), renal insufficiency (serum creatinine ≥ 2 mg/dL), anemia (hemoglobin value below the lower limit of normal by >2 g/dL or hemoglobin value < 10 g/dL), and lytic bone lesions (CRAB features) that can be attributed to the plasma cell proliferative disorder (7). SMM was defined by the presence of serum M-protein (≥ 3 g/dL) and/or clonal bone marrow plasma cells (≥ 10%) and the absence of CRAB features, clinically.

Recently, 3 clinical subtypes of MGUS have been defined on the basis of the type of immunoglobulin involved—non-IgM MGUS, IgM MGUS, and light-chain MGUS (8) and are characterized by a unique natural history in each subtype.

In his initial description of monoclonal gammopathy, Jan Waldenström speculated that "essential benign hypergammaglobulinemia" was more common than multiple myeloma (2). We now know that indeed MGUS is the most prevalent plasma cell disorder (9). In a large population-based study in Olmsted County, Minnesota, Kyle and colleagues analyzed serum samples of more than 75% of residents, 50 years or older, within the county (9). They identified MGUS in 3.2% of 21,463 patients tested. While the overall prevalence was noted to be 3.2%, there was a significant age-dependent increase in both sexes with the prevalence among persons aged 80 years or older 4 times as high as among those aged 50 to 59 years. In a subsequent study on a majority of the same patients from Olmsted County, Dispenzieri and colleagues used the free light-chain assay (FREELITE) and showed that 0.8% of people aged more than 50 years had light-chain MGUS. The total MGUS prevalence including the light-chain MGUS cases was noted at 4.2% (10). Other studies from across the globe have shown significant variation in the prevalence of MGUS (Table 1). The prevalence seems to be low in Mexico and among the Asian countries, based on studies from Japan, Taiwan, and Thailand (11–14), while much higher in Africans and African-Americans (15, 16).

Table 1.

Worldwide incidence of MGUS

Study populationAgeNo. of participantsPrevalence of MGUS (%) with 95% CI
Olmsted County, MN-2006 (9) >50 21,463 3.2 (3.0–3.5) 
Olmsted County, MN-2010 (10) >50 18,357 4.2 (3.9–4.5) 
Nagasaki City, Japan (11) >42 52,802 2.1 (1.9–2.2) 
Thailand (13) >50 3,260 2.3 (1.8–28) 
Ghana (15) 50–74 917 5.84 (4.27–7.4) 
Udine, Italy (64) 18–67 8,197 1.3 
Finistere, Francea (19) >30 30, 279 1.1 
General Hospital, Italya (65) — 102,000 0.7 
Swedena (66) >25 6,995 0.9 
Study populationAgeNo. of participantsPrevalence of MGUS (%) with 95% CI
Olmsted County, MN-2006 (9) >50 21,463 3.2 (3.0–3.5) 
Olmsted County, MN-2010 (10) >50 18,357 4.2 (3.9–4.5) 
Nagasaki City, Japan (11) >42 52,802 2.1 (1.9–2.2) 
Thailand (13) >50 3,260 2.3 (1.8–28) 
Ghana (15) 50–74 917 5.84 (4.27–7.4) 
Udine, Italy (64) 18–67 8,197 1.3 
Finistere, Francea (19) >30 30, 279 1.1 
General Hospital, Italya (65) — 102,000 0.7 
Swedena (66) >25 6,995 0.9 

aThese older studies did not use the modern definition of MGUS.

In a study of 4 million veterans in Veterans Affairs hospitals across the nation, Landgren and colleagues showed that the age-adjusted prevalence rate of MGUS was 3-fold higher for African-American patients than for white patients (16). This study also showed that the rate of progression to multiple myeloma was similar among whites and African-Americans.

The possible role of genetic factors in MGUS is suggested by the racial and ethnic differences mentioned above and also the increased prevalence of MGUS in blood relatives of persons with lymphoproliferative and plasma cell disorders. First-degree relatives of multiple myeloma and MGUS patients were found to have a 2.6-fold risk of MGUS as compared with the general population (17).

Environmental factors have been evaluated, and a study showed that exposure to pesticides increases the risk of MGUS (18). In a French study, the prevalence of monoclonal gammopathy was higher in the rural areas as compared with urban areas (19). A link between obesity and MGUS has also been suggested (20).

MGUS consistently precedes multiple myeloma

Since the early description of MGUS, it was known that some cases of MGUS progressed to symptomatic myeloma, but it was not clear whether all cases of myeloma are preceded by MGUS. In a study of more than 77,000 individuals, 55 to 74 years of age, from a cancer screening trial, Landgren and colleagues found 71 patients who developed multiple myeloma (5). The study showed that patients who eventually developed multiple myeloma consistently had MGUS in the years preceding development of multiple myeloma. A second study by Weiss and colleagues found a monoclonal gammopathy in 27 of 30 patients (90%) in sera 2 or more years before the diagnosis of myeloma (6).

Studies looking at the epigenetic, genetic, transcriptional, and phenotypic changes within the transforming plasma cell as well as the microenvironment have improved our understanding of this process. The process of transformation, from a normal plasma cell to the premalignant MGUS/SMM state and finally to symptomatic multiple myeloma, involves several oncogenic events within the plasma cell as well as in the bone marrow microenvironment (Figure 1).

Figure 1.

Biology of myelomagenesis. Progression from a normal postgerminal center B cell to symptomatic myeloma involves a series of genetic and phenotypic changes. Early genetic events can be categorized into 2 major types, hyperdiploid type and nonhyperdiploid type. These are unified by cyclin D dysregulation (30). The role of 13q seems to be less certain and it may be an early or late event. Secondary genetic changes include Ras mutations, NF-κB pathway–activating mutations, and inactivating mutations of p53, PTEN, or RB pathways (1). Several other genetic changes also occur along the way, including secondary translocations, increase in copy number abnormalities, HOXA9 overexpression, and Myc upregulation (21, 36). The end result of these changes includes a clonal expansion of abnormal plasma cells that occupy bone marrow stem cell niches (45). Lytic bone lesions are seen due to increased osteoclast activation resulting from RANK-L signaling and osteoblast inhibition from DKK1 activity (41, 44). Immune evasion, paracrine, and autocrine signaling mediated by cytokine and growth factors are important final steps (46, 51).

Figure 1.

Biology of myelomagenesis. Progression from a normal postgerminal center B cell to symptomatic myeloma involves a series of genetic and phenotypic changes. Early genetic events can be categorized into 2 major types, hyperdiploid type and nonhyperdiploid type. These are unified by cyclin D dysregulation (30). The role of 13q seems to be less certain and it may be an early or late event. Secondary genetic changes include Ras mutations, NF-κB pathway–activating mutations, and inactivating mutations of p53, PTEN, or RB pathways (1). Several other genetic changes also occur along the way, including secondary translocations, increase in copy number abnormalities, HOXA9 overexpression, and Myc upregulation (21, 36). The end result of these changes includes a clonal expansion of abnormal plasma cells that occupy bone marrow stem cell niches (45). Lytic bone lesions are seen due to increased osteoclast activation resulting from RANK-L signaling and osteoblast inhibition from DKK1 activity (41, 44). Immune evasion, paracrine, and autocrine signaling mediated by cytokine and growth factors are important final steps (46, 51).

Close modal

A recent study using single-nucleotide polymorphism-based arrays compared MGUS, SMM, and multiple myeloma samples (21). They found copy number abnormalities in all stages. The incidence of genomic imbalance increased from a median of 5 per case for MGUS to 7.5 per case for SMM and 12 per case for multiple myeloma. The study also noted certain genomic changes that were apparently exclusive to multiple myeloma, including 11q and 21q gains and 16q and 22q deletions. Interestingly, the study found these abnormalities in a small subclone in patients with MGUS indicating that most, if not all, of the chromosomal changes may be already present at the MGUS state.

Several studies have found varying rates of the different translocations between MGUS, SMM, and multiple myeloma (Table 2; refs. 22–27). The rate of t(11;14) seems to be uniform from MGUS to multiple myeloma, but there is some discrepancy in the rate of the other IgH translocations—t(4;14) and t(14;16), as well as del13q. Some studies have suggested that the incidence of these changes increases from MGUS to multiple myeloma, whereas other studies have indicated that the rate is the same among patients with MGUS and multiple myeloma. Some studies have suggested that 13q deletion may be an early event, whereas others point to del13q as the next step in the pathogenesis of myeloma, especially following IgH translocations (25, 27). The presence of these translocations in MGUS and SMM does not seem to affect the rate of progression in either condition, whereas in multiple myeloma some of these translocations are associated with adverse outcomes (24, 28, 29). A recent study has shown that there is a clonal expansion of these genetically abnormal plasma cells from MGUS to SMM and multiple myeloma (26). IgH translocations and 13q deletion were seen in a higher proportion of plasma cells in SMM compared with MGUS and in multiple myeloma compared with SMM.

Table 2.

Common cytogenetic abnormalities and their incidence in MGUS, SMM, and multiple myeloma (22–27)

Cytogenetic abnormalityInvolved oncogeneMGUS%SMM%Multiple myeloma%
IgH translocations See below 40%–50% 40%–50% 50%–70% 
 t(11;14)(q13;q32) CCND1 (cyclin D1) 10%–25% 10%–25% 15% 
 t(4;14)(p16;q32) FGFR3 and MMSET 2%–9% 3%–13% 10%–15% 
 t(14;16)(q32;q23) C-MAF 2%–5% 2%–5% 2%–5% 
Other IgH translocations 
 t(6;14)(p21;q32), t(14;20)(q32;q11) etc. CCND3 (cyclin D3), MAFB, etc. 6%–10% 1%–10% 10% 
13q deletion Unknown 25%–50% 35%–50% 40%–50% 
Hyperdiploidy Unknown 40%–50% 40%–50% 40%–50% 
Cytogenetic abnormalityInvolved oncogeneMGUS%SMM%Multiple myeloma%
IgH translocations See below 40%–50% 40%–50% 50%–70% 
 t(11;14)(q13;q32) CCND1 (cyclin D1) 10%–25% 10%–25% 15% 
 t(4;14)(p16;q32) FGFR3 and MMSET 2%–9% 3%–13% 10%–15% 
 t(14;16)(q32;q23) C-MAF 2%–5% 2%–5% 2%–5% 
Other IgH translocations 
 t(6;14)(p21;q32), t(14;20)(q32;q11) etc. CCND3 (cyclin D3), MAFB, etc. 6%–10% 1%–10% 10% 
13q deletion Unknown 25%–50% 35%–50% 40%–50% 
Hyperdiploidy Unknown 40%–50% 40%–50% 40%–50% 

NOTE: Studies investigating the cytogenetic abnormalities in MGUS and SMM are limited by the small number of subjects in each group as well as by technical limitations in the ability to select abnormal plasma cells using CD138+ sorting methods. The numbers of subjects in each study are as follows: 123 high-risk SMM (22); 62 MGUS and 10 SMM (24); 189 MGUS, 127 SMM, and 400 multiple myeloma (25); 90 MGUS, 102 high-risk SMM, and 373 multiple myeloma (26); and 147 MGUS, 39 SMM, and 669 multiple myeloma (27).

Cyclin D overexpression as a unifying early event

Using gene expression analyses, overexpression of the cyclin D genes seems to be one possible unifying event that is seen in almost all multiple myeloma cases with or without an immunoglobulin translocation as compared with normal plasma cells (30). IgH translocations directly dysregulate cyclin D1 or D3 [t(11;14) and t(6;14), respectively] and the C-MAF or MAFB transcription factors dysregulate cyclin D2 [t(14;16) and t(14;20); ref. 31]. In this study, 12 MGUS samples had a similar pattern of cyclin D dysregulation despite a lower proliferative index, suggesting that cyclin D perturbation may indeed be an early and unifying event in plasma cell dyscrasias.

MicroRNAs (miRNA) are single-stranded RNA molecules that regulate gene expression posttranscriptionally and are being implicated in a large number of cancers (32). A study comparing miRNA profiles of normal plasma cells (PC), MGUS, SMM, and multiple myeloma found overexpression of mir-21, mir-106∼25, and mir181a, and mir-181b in MGUS and multiple myeloma compared to normal PCs (33). The study also found mir-32 and mir17∼92 clusters to be upregulated only in multiple myeloma and not in any other forms. mir-17∼92 and mir-106∼25 are known to have a role in B-cell development as well as B-cell lymphomas targeting PTEN, E2F1, Bcl-2, and BIM (32, 33). The mir-17 cluster has been shown to upregulated by the transcription factor c-Myc, which is considered a late event in myeloma progression (34, 35).

Cytogenetic and gene expression studies in myeloma precursor disease are limited by technical limitations in the ability to sort out the abnormal plasma cells for evaluation. The ratio of abnormal cells to normal plasma cells will be lower in a MGUS sample than in a multiple myeloma sample if CD 138+ sorting is used.

With the advances in deep sequencing techniques, we are now able to study the whole genome of myeloma cells and compare that with the normal human genome. Several studies using these techniques are beginning to shift our understanding of the pathogenesis of myeloma. These studies have revealed that the previously held dogma that cancer progression occurred through a linear cumulative acquisition of mutational changes may be too simplistic. A new model of clonal heterogeneity is beginning to emerge (36). In this model, various subclones exist in a dynamic equilibrium and, over time, the subclonal populations ebb and flow under environmental evolutionary pressures with alternating dominance of various subclones (37–40). Most of these studies were done on patients with symptomatic multiple myeloma and as similar studies involving MGUS and SMM emerge, our understanding of the pathogenesis of myeloma from these precursor states will continue to evolve.

Role of microenvironment in progression of MGUS to multiple myeloma

The transition from MGUS to multiple myeloma involves changes to the complex interaction with the microenvironment along with the genetic changes described above. In multiple myeloma, there is an upregulation of osteoblast RANK-L expression and a decrease in osteoprotegerin (OPG), a decoy for RANK-L that inhibits osteoclast differentiation. This increases the ratio of RANK-L/OPG, which leads to osteoclast activation and development of the characteristic lytic lesions (41). Interestingly, although lytic bone lesions are not seen in MGUS, the RANK-L/OPG ratio is higher in MGUS subjects and they are at a higher risk of fractures than healthy controls (42, 43). The role of other pathways including Wnt/β-catenin involving activation of FRZB and Dickkopf 1 (DKK1), a Wnt inhibitor, is also being studied (44). The homing of multiple myeloma cells to bone marrow stromal cell niches is essential for their survival. A recent study suggests that these bone marrow stromal cell niches are limited, and a progressive competition and replacement of normal bone marrow cells by clonal plasma cells is associated with more advanced disease (45). The evasion of the immune system is an important step in the progression from MGUS to multiple myeloma. Oligoclonal T-cell expansion can be found in both MGUS and multiple myeloma with lower disease burden states showing a more robust T-cell expansion (46, 47). A defect in the T-cell function is seen during progression from MGUS to multiple myeloma (48, 49). Several cytokines and growth factors including interleukin (IL)-6, SDF-1, macrophage inflammatory protein (MIP)-1α, insulin-like growth factor (IGF)-I, VEGF, and hepatocyte growth factor (HGF) have been implicated in myeloma pathogenesis and transition from MGUS (50, 51).

It is clear from the description above that there are no major genetic differences between the plasma cells in MGUS and multiple myeloma. Since the prevalence of MGUS/SMM is much higher than multiple myeloma, and several people with precursor disease will never progress to symptomatic myeloma in their lifetime, it is important to identify patients that are at greatest risk for progression to myeloma. Several studies have tried to predict subgroups of precursor disease at higher risk of progression using different tools.

Size of M protein

In 1,384 patients with MGUS, the risk of progression to multiple myeloma or a related disorder after 20 years was 14% with an initial protein level of 0.5 g/dL or less, 25% for 1.5 g/dL, 41% for 2 g/dL, 49% for 2.5 g/dL, and 64% for 3 g/dL (52). A progressive increase in the M-protein in the first year of follow-up (evolving MGUS) has also shown to be prognostic (53).

Type of immunoglobulin

Patients with MGUS with IgM or IgA monoclonal protein have an increased risk of progression to disease as compared with patients with IgG protein (52).

Serum-free light-chain ratio and the Mayo Clinic model

In a study of 1,148 patients with MGUS, Rajkumar and colleagues showed that an abnormal free light-chain (FLC) assay was an independent risk factor for progression from MGUS to multiple myeloma (54). For patients with MGUS, a non-IgG isotype, M-protein concentration more than 1.5 g/dL, and an abnormal FLC ratio are considered adverse prognostic factors. At 20 years, the risk of progression in patients with 0, 1, 2, and 3 risk factors is 5%, 21%, 37%, and 58%, respectively (Table 3). For patients with SMM, an M-protein ≥ 3 g/dL, an FLC ratio outside the range of 0.125 to 8, and ≥ 10% plasma cells in the bone marrow are considered as adverse factors in this model (55, 56). The 5-year rate of progression in patients with 1, 2, and 3 risk factors was 25%, 51%, and 76%, respectively (Table 5). Recently, Rajkumar and colleagues have proposed that SMM with more than 60% plasma cells progresses to multiple myeloma within 2 years in 95% cases and should be treated at diagnosis even in the absence of symptoms (57).

Table 3.

Risk stratification model for MGUS using Mayo Clinic model (54)

ModelNo. of risk factors20-year progression, %Relative risk
Mayo Clinic model 
Risk factors: 21 5.4 
M-protein ≥1.5 g/dL; 37 10.1 
non-IgG MGUS; 58 20.8 
FLC ratio <0.26 or >1.65 Total 20 NA 
ModelNo. of risk factors20-year progression, %Relative risk
Mayo Clinic model 
Risk factors: 21 5.4 
M-protein ≥1.5 g/dL; 37 10.1 
non-IgG MGUS; 58 20.8 
FLC ratio <0.26 or >1.65 Total 20 NA 

Abbreviation: NA, not applicable.

Flow cytometry and the Spanish model

Perez-Persona and colleagues used immunophenotyping with multiparameter flow cytometry to identify aberrant plasma cell (aPC) in the bone marrow of 407 MGUS and 93 SMM patients (58). A ratio of aPC/BMPC of more than 95% was shown to be an independent risk factor for progression in both MGUS and SMM. In a multivariate analysis, DNA aneuploidy (hypo- or hyperdiploidy) was also noted to be a prognostic factor in MGUS and was combined with aPC/BMPC of more than 95% to form a prognostic index. The presence of 0, 1, or 2 of these factors was associated with a progression risk of 2%, 10%, and 46%, respectively, in MGUS (Table 4). The analysis identified immunoparesis as an independent prognostic factor in SMM. Using aPC/BMPC of more than 95% and immunoparesis as the 2 factors the study found 5-year risk of 4%, 46%, and 72% in patients with 0, 1, or 2 factors, respectively (Table 6).

Table 4.

Risk stratification model for MGUS using Spanish (PETHEMA) model (58)

ModelNo. of risk factors5-year progression, %Relative risk
Spanish (PETHEMA) 
model 10 
Risk factors: ≥95% 46 23 
aPC; DNA aneuploidy Total 8.5 NA 
ModelNo. of risk factors5-year progression, %Relative risk
Spanish (PETHEMA) 
model 10 
Risk factors: ≥95% 46 23 
aPC; DNA aneuploidy Total 8.5 NA 

Abbreviation: NA, not applicable.

Table 5.

Risk stratification model for SMM using Mayo Clinic model (56)

ModelNo. of risk factors5-year progression, %Relative risk
Mayo Clinic model 25 
Risk factors: M-protein 51 2.0 
≥3 g/dL; ≥10% bone 76 3.0 
marrow plasma cells; FLC ratio <0.125 or >8 Total 51 NA 
ModelNo. of risk factors5-year progression, %Relative risk
Mayo Clinic model 25 
Risk factors: M-protein 51 2.0 
≥3 g/dL; ≥10% bone 76 3.0 
marrow plasma cells; FLC ratio <0.125 or >8 Total 51 NA 

Abbreviation: NA, not applicable.

Table 6.

Risk stratification model for MGUS using Spanish (PETHEMA) model (58)

ModelNo. of risk factors5-year progression, %Relative risk
Spanish (PETHEMA) 
model 46 11.5 
Risk factors: ≥95% 72 18 
aPC; immunoparesis Total 46 NA 
ModelNo. of risk factors5-year progression, %Relative risk
Spanish (PETHEMA) 
model 46 11.5 
Risk factors: ≥95% 72 18 
aPC; immunoparesis Total 46 NA 

Abbreviation: NA, not applicable.

Other prognostic factors

Other studies have identified various factors that are prognostic for progression to symptomatic disease. Cesana and colleagues proposed a prognostic index for MGUS based on the presence of BMPC infiltrate more than 5%, presence of Bence–Jones proteinuria, polyclonal serum Ig reduction, and erythrocyte sedimentation rate (ESR) and showed that these features were associated with higher rate of transformation to myeloma (59). Whole body MRI has been shown to be more sensitive to a skeletal survey in detecting focal lesions and the presence of such focal lesions were an adverse prognostic factor for progression to multiple myeloma (60). An earlier study showed that an abnormal MRI was associated with a shorter time to progression compared with a normal MRI (1.5 vs. 5 years) in SMM (61). Immunoparesis (suppression of uninvolved immunoglobulins) has been shown to be prognostic in SMM but its value in MGUS is less clear (52, 55, 58, 59). A recent study also looked at each isotype-specific heavy and light chain in MGUS and suppression of the uninvolved HLC (e.g., IgG-λ suppression in an IgG-κ MGUS patient) was shown to be prognostic for progression (62). In a study of 325 patients with MGUS, the presence of circulating plasma cells was shown to be prognostic for progression (HR, 2.1; P, 0.03) compared with patients who did not have any circulating plasma cells (63). Age, sex, race, and chromosomal translocations are not prognostic factors for progression from precursor disease to symptomatic myeloma.

Despite all the recent advances in the treatment of multiple myeloma, it remains an incurable disease. The myeloma precursor conditions provide an opportunity to be able to intervene at an earlier stage to either delay or prevent progression altogether. Several trials have looked at early intervention in an attempt to change the natural history of multiple myeloma. These trials have produced mixed results and other than the Programa para el Estudio de la Terapeutica en Hemopatias Malignas (PETHEMA) trial have not shown an improvement in overall survival (Table 7). A longer follow-up will be necessary to ascertain whether the benefits from early treatment are sustained without long-term adverse effects. Several other studies testing novel drugs are ongoing for patients with SMM (Table 8). The current standard of care based on the IMWG recommendations (Supplementary Fig. S1) is to monitor patients with MGUS or SMM until they progress to multiple myeloma before starting treatment (7).

Table 7.

Completed intervention studies for myeloma precursor disease

Drug/sReferenceComments
Melphalan and prednisone (67) Fifty patients with SMM were randomized to receive melphalan and prednisone immediately upon diagnosis or at the time of disease progression. The study found no difference in the response rate or overall survival in the 2 groups. 
Thalidomide (68) A small single-arm study of 16 subjects showed that thalidomide produced a PR or better in 6 of 16 subjects. 
Thalidomide and pamidronate (69) In this phase I study, 76 patients with SMM were administered thalidomide daily and pamidronate monthly. The study had a 50% discontinuation rate within the first 2 years due to adverse effects. Unexpectedly, the study found that attaining a PR was associated with a shorter time to salvage therapy for disease progression. 
Zoledronic acid (70) A prospective, open label, randomized trial comparing zoledronic acid versus observation in 163 patients with SMM. Monthly zolderonic acid for 1 year in patients with SMM reduced the development of skeletal-related events (SRE) at the time of progression (55.5% vs. 78.3%; P = 0.041) but did not affect the time to progression to symptomatic disease (67 vs. 59 months; P = 0.83). 
Curcumin (71) In a double-blind placebo-controlled study, curcumin was shown to have a modest effect in decreasing the FLC ratio among a quarter of MGUS and SMM subjects, but the effect on progression to symptomatic disease was not reported. 
Lenalidomide and dexamethasone (72) A phase III PETHEMA trial of high-risk SMM randomized patients to induction with lenalidomide and dexamethasone followed by lenalidomide maintenance versus no treatment. After a median follow-up of 32 months, 9 (15%) patients in the treatment arm and 37 (59%) patients in the observation arm had progressed (HR, 6.0; 95% CI, 2.9–12.6; P < 0.0001). The estimated 3-year overall survival was 93% in the treatment arm and 76% in the observation arm (P = 0.04). 
Drug/sReferenceComments
Melphalan and prednisone (67) Fifty patients with SMM were randomized to receive melphalan and prednisone immediately upon diagnosis or at the time of disease progression. The study found no difference in the response rate or overall survival in the 2 groups. 
Thalidomide (68) A small single-arm study of 16 subjects showed that thalidomide produced a PR or better in 6 of 16 subjects. 
Thalidomide and pamidronate (69) In this phase I study, 76 patients with SMM were administered thalidomide daily and pamidronate monthly. The study had a 50% discontinuation rate within the first 2 years due to adverse effects. Unexpectedly, the study found that attaining a PR was associated with a shorter time to salvage therapy for disease progression. 
Zoledronic acid (70) A prospective, open label, randomized trial comparing zoledronic acid versus observation in 163 patients with SMM. Monthly zolderonic acid for 1 year in patients with SMM reduced the development of skeletal-related events (SRE) at the time of progression (55.5% vs. 78.3%; P = 0.041) but did not affect the time to progression to symptomatic disease (67 vs. 59 months; P = 0.83). 
Curcumin (71) In a double-blind placebo-controlled study, curcumin was shown to have a modest effect in decreasing the FLC ratio among a quarter of MGUS and SMM subjects, but the effect on progression to symptomatic disease was not reported. 
Lenalidomide and dexamethasone (72) A phase III PETHEMA trial of high-risk SMM randomized patients to induction with lenalidomide and dexamethasone followed by lenalidomide maintenance versus no treatment. After a median follow-up of 32 months, 9 (15%) patients in the treatment arm and 37 (59%) patients in the observation arm had progressed (HR, 6.0; 95% CI, 2.9–12.6; P < 0.0001). The estimated 3-year overall survival was 93% in the treatment arm and 76% in the observation arm (P = 0.04). 
Table 8.

Currently ongoing intervention studies for precursor disease (73)

DrugTarget/MOAStudy phaseComments
IPH2101 Anti-KIR monoclonal Ab Single-arm study assessing response of drug in SMM and measuring toxicity. 
BHQ880 Anti-Dickkopf (DKK1) Ab Single-arm study assessing response of drug in high-risk SMM. 
Elotuzumab Anti-CS1 monoclonal Ab Study looking at association between NK cell status and efficacy of elotuzumab monotherapy in high-risk SMM. 
Slituximab Anti-IL6 monoclonal Ab Randomized, blinded, placebo-controlled trial looking at one-year progression-free survival (PFS) in high-risk SMM. 
MLN 9708 and dexamethasone Proteasome inhibitor and steroid A single-arm study in high-risk SMM with response rate as primary outcome and PFS and duration of response as secondary outcomes. 
Lenalidomide IMiD A phase III randomized Eastern Cooperative Oncology Group (ECOG) trial of lenalidomide vs observation in high-risk SMM. 
Bortezomib Proteasome inhibitor Study looking at the effects of low-dose (0.7 mg/m2 i.v.) bortezomib on bone formation and natural history in SMM. 
Carflizomib, lenalidomide, and dexamethasone Proteasome inhibitor, IMiD, and steroid Single-arm study in high-risk SMM assessing the safety and response rate of the triple-drug combination in SMM. 
DrugTarget/MOAStudy phaseComments
IPH2101 Anti-KIR monoclonal Ab Single-arm study assessing response of drug in SMM and measuring toxicity. 
BHQ880 Anti-Dickkopf (DKK1) Ab Single-arm study assessing response of drug in high-risk SMM. 
Elotuzumab Anti-CS1 monoclonal Ab Study looking at association between NK cell status and efficacy of elotuzumab monotherapy in high-risk SMM. 
Slituximab Anti-IL6 monoclonal Ab Randomized, blinded, placebo-controlled trial looking at one-year progression-free survival (PFS) in high-risk SMM. 
MLN 9708 and dexamethasone Proteasome inhibitor and steroid A single-arm study in high-risk SMM with response rate as primary outcome and PFS and duration of response as secondary outcomes. 
Lenalidomide IMiD A phase III randomized Eastern Cooperative Oncology Group (ECOG) trial of lenalidomide vs observation in high-risk SMM. 
Bortezomib Proteasome inhibitor Study looking at the effects of low-dose (0.7 mg/m2 i.v.) bortezomib on bone formation and natural history in SMM. 
Carflizomib, lenalidomide, and dexamethasone Proteasome inhibitor, IMiD, and steroid Single-arm study in high-risk SMM assessing the safety and response rate of the triple-drug combination in SMM. 

The recent advances in our knowledge about MGUS and SMM have helped us better understand the pathogenesis of myeloma. It seems that myeloma evolves from a precursor state in almost all cases. We do not completely understand the multistep process from the precursor state to myeloma, but studies including whole genome sequencing will continue to help in improving our understanding of this process. The process of transformation may not be linear acquisition of changes but rather a branched heterogeneous process. Clinical features that are prognostic of rapid transformation to symptomatic disease have been identified, but no specific molecular markers have been identified. Intervening at the precursor state provides a unique opportunity to alter the natural history of the disease. A limitation is that a vast majority of patients with precursor disease, especially low-risk MGUS, will never progress to myeloma in their lifetime and treating these patients is not only unnecessary but may be potentially harmful. The challenge is to identify a subset of patients with the precursor state that would definitely progress to myeloma and in whom interventions will have a meaningful impact. As our understanding of the molecular and genetic processes improves, these studies will guide the selection of high-risk patients more appropriately and ultimately direct a tailored management strategy.

I.M. Ghobrial is a consultant/advisory board member of Novartis, Onyx, Millennium, and Bristol-Myers Squibb. No potential conflicts of interest were disclosed by the other author.

Conception and design: A. Agarwal, I.M. Ghobrial

Development of methodology: A. Agarwal, I.M. Ghobrial

Acquisition of data (provided animals, acquired and managed patients, provided facilities, etc.): A. Agarwal

Writing, review, and/or revision of the manuscript: A. Agarwal, I.M. Ghobrial

I.M. Ghobrial is supported in part by grants R01CA125690, R01CA133799, and R01CA154648.

1.
Anderson
KC
,
Carrasco
RD
. 
Pathogenesis of myeloma
.
Annu Rev Pathol
2011
;
6
:
249
74
.
2.
Waldenstrom
J
. 
Studies on conditions associated with disturbed gamma globulin formation (gammopathies)
.
Harvey Lect
1960
;
56
:
211
31
.
3.
Kyle
RA
. 
Monoclonal gammopathy of undetermined significance. Natural history in 241 cases
.
Am J Med
1978
;
64
:
814
26
.
4.
Kyle
RA
,
Greipp
PR
. 
Smoldering multiple myeloma
.
N Engl J Med
1980
;
302
:
1347
9
.
5.
Landgren
O
,
Kyle
RA
,
Pfeiffer
RM
,
Katzmann
JA
,
Caporaso
NE
,
Hayes
RB
, et al
Monoclonal gammopathy of undetermined significance (MGUS) consistently precedes multiple myeloma: a prospective study
.
Blood
2009
;
113
:
5412
7
.
6.
Weiss
BM
,
Abadie
J
,
Verma
P
,
Howard
RS
,
Kuehl
WM
. 
A monoclonal gammopathy precedes multiple myeloma in most patients
.
Blood
2009
;
113
:
5418
22
.
7.
Kyle
RA
,
Durie
BG
,
Rajkumar
SV
,
Landgren
O
,
Blade
J
,
Merlini
G
, et al
Monoclonal gammopathy of undetermined significance (MGUS) and smoldering (asymptomatic) multiple myeloma: IMWG consensus perspectives risk factors for progression and guidelines for monitoring and management
.
Leukemia
2010
;
24
:
1121
7
.
8.
Rajkumar
SV
,
Kyle
RA
,
Buadi
FK
. 
Advances in the diagnosis, classification, risk stratification, and management of monoclonal gammopathy of undetermined significance: implications for recategorizing disease entities in the presence of evolving scientific evidence
.
Mayo Clin Proc
2010
;
85
:
945
8
.
9.
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
.
10.
Dispenzieri
A
,
Katzmann
JA
,
Kyle
RA
,
Larson
DR
,
Melton
LJ
 III
,
Colby
CL
, et al
Prevalence and risk of progression of light-chain monoclonal gammopathy of undetermined significance: a retrospective population-based cohort study
.
Lancet
2010
;
375
:
1721
8
.
11.
Iwanaga
M
,
Tagawa
M
,
Tsukasaki
K
,
Kamihira
S
,
Tomonaga
M
. 
Prevalence of monoclonal gammopathy of undetermined significance: study of 52,802 persons in Nagasaki City, Japan
.
Mayo Clin Proc
2007
;
82
:
1474
9
.
12.
Lee
LN
,
Jan
IS
,
Tien
HF
,
Lin
JS
,
Lo
SC
,
Cheng
WC
. 
Laboratory and clinical characterization of monoclonal gammopathy in Taiwanese
.
J Formos Med Assoc
2002
;
101
:
91
7
.
13.
Watanaboonyongcharoen
P
,
Nakorn
TN
,
Rojnuckarin
P
,
Lawasut
P
,
Intragumtornchai
T
. 
Prevalence of monoclonal gammopathy of undetermined significance in Thailand
.
Int J Hematol
2012
;
95
:
176
81
.
14.
Ruiz-Delgado
GJ
,
Ruiz-Arguelles
GJ
. 
Genetic predisposition for monoclonal gammopathy of undetermined significance
.
Mayo Clin Proc
2008
;
83
:
601
2
.
15.
Landgren
O
,
Katzmann
JA
,
Hsing
AW
,
Pfeiffer
RM
,
Kyle
RA
,
Yeboah
ED
, et al
Prevalence of monoclonal gammopathy of undetermined significance among men in Ghana
.
Mayo Clin Proc
2007
;
82
:
1468
73
.
16.
Landgren
O
,
Gridley
G
,
Turesson
I
,
Caporaso
NE
,
Goldin
LR
,
Baris
D
, et al
Risk of monoclonal gammopathy of undetermined significance (MGUS) and subsequent multiple myeloma among African American and white veterans in the United States
.
Blood
2006
;
107
:
904
6
.
17.
Vachon
CM
,
Kyle
RA
,
Therneau
TM
,
Foreman
BJ
,
Larson
DR
,
Colby
CL
, et al
Increased risk of monoclonal gammopathy in first-degree relatives of patients with multiple myeloma or monoclonal gammopathy of undetermined significance
.
Blood
2009
;
114
:
785
90
.
18.
Landgren
O
,
Kyle
RA
,
Hoppin
JA
,
Beane
Freeman LE
,
Cerhan
JR
,
Katzmann
JA
, et al
Pesticide exposure and risk of monoclonal gammopathy of undetermined significance in the Agricultural Health Study
.
Blood
2009
;
113
:
6386
91
.
19.
Saleun
JP
,
Vicariot
M
,
Deroff
P
,
Morin
JF
. 
Monoclonal gammopathies in the adult population of Finistere, France
.
J Clin Pathol
1982
;
35
:
63
8
.
20.
Landgren
O
,
Rajkumar
SV
,
Pfeiffer
RM
,
Kyle
RA
,
Katzmann
JA
,
Dispenzieri
A
, et al
Obesity is associated with an increased risk of monoclonal gammopathy of undetermined significance among black and white women
.
Blood
2010
;
116
:
1056
9
.
21.
Lopez-Corral
L
,
Sarasquete
ME
,
Bea
S
,
Garcia-Sanz
R
,
Mateos
MV
,
Corchete
LA
, et al
SNP-based mapping arrays reveal high genomic complexity in monoclonal gammopathies, from MGUS to myeloma status
.
Leukemia
2012
;
26
:
2521
9
.
22.
Lopez-Corral
L
,
Mateos
MV
,
Corchete
LA
,
Sarasquete
ME
,
de la Rubia
J
,
de Arriba
F
, et al
Genomic analysis of high risk smoldering multiple myeloma
.
Haematologica
2012
;
97
:
1439
43
.
23.
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
.
24.
Fonseca
R
,
Bailey
RJ
,
Ahmann
GJ
,
Rajkumar
SV
,
Hoyer
JD
,
Lust
JA
, et al
Genomic abnormalities in monoclonal gammopathy of undetermined significance
.
Blood
2002
;
100
:
1417
24
.
25.
Chiecchio
L
,
Dagrada
GP
,
Ibrahim
AH
,
Dachs
Cabanas E
,
Protheroe
RK
,
Stockley
DM
, et al
Timing of acquisition of deletion 13 in plasma cell dyscrasias is dependent on genetic context
.
Haematologica
2009
;
94
:
1708
13
.
26.
Lopez-Corral
L
,
Gutierrez
NC
,
Vidriales
MB
,
Mateos
MV
,
Rasillo
A
,
Garcia-Sanz
R
, et al
The progression from MGUS to smoldering myeloma and eventually to multiple myeloma involves a clonal expansion of genetically abnormal plasma cells
.
Clin Cancer Res
2011
;
17
:
1692
700
.
27.
Avet-Loiseau
H
,
Facon
T
,
Grosbois
B
,
Magrangeas
F
,
Rapp
MJ
,
Harousseau
JL
, et al
Oncogenesis of multiple myeloma: 14q32 and 13q chromosomal abnormalities are not randomly distributed, but correlate with natural history, immunological features, and clinical presentation
.
Blood
2002
;
99
:
2185
91
.
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.
Gertz
MA
,
Lacy
MQ
,
Dispenzieri
A
,
Greipp
PR
,
Litzow
MR
,
Henderson
KJ
, et al
Clinical implications of t(11;14)(q13;q32), t(4;14)(p16.3;q32), and -17p13 in myeloma patients treated with high-dose therapy
.
Blood
2005
;
106
:
2837
40
.
30.
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
.
31.
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
.
32.
Visone
R
,
Croce
CM
. 
MiRNAs and cancer
.
Am J Pathol
2009
;
174
:
1131
8
.
33.
Pichiorri
F
,
Suh
SS
,
Ladetto
M
,
Kuehl
M
,
Palumbo
T
,
Drandi
D
, et al
MicroRNAs regulate critical genes associated with multiple myeloma pathogenesis
.
Proc Natl Acad Sci U S A
2008
;
105
:
12885
90
.
34.
O'Donnell
KA
,
Wentzel
EA
,
Zeller
KI
,
Dang
CV
,
Mendell
JT
. 
c-Myc-regulated microRNAs modulate E2F1 expression
.
Nature
2005
;
435
:
839
43
.
35.
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
.
36.
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
.
37.
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
.
38.
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
.
39.
Walker
BA
,
Wardell
CP
,
Melchor
L
,
Hulkki
S
,
Potter
NE
,
Johnson
DC
, et al
Intraclonal heterogeneity and distinct molecular mechanisms characterize the development of t(4;14) and t(11;14) myeloma
.
Blood
2012
;
120
:
1077
86
.
40.
Bahlis
NJ
. 
Darwinian evolution and tiding clones in multiple myeloma
.
Blood
2012
;
120
:
927
8
.
41.
Roodman
GD
. 
Pathogenesis of myeloma bone disease
.
Leukemia
2009
;
23
:
435
41
.
42.
Politou
M
,
Terpos
E
,
Anagnostopoulos
A
,
Szydlo
R
,
Laffan
M
,
Layton
M
, et al
Role of receptor activator of nuclear factor-kappa B ligand (RANKL), osteoprotegerin and macrophage protein 1-alpha (MIP-1a) in monoclonal gammopathy of undetermined significance (MGUS)
.
Br J Haematol
2004
;
126
:
686
9
.
43.
Kristinsson
SY
,
Tang
M
,
Pfeiffer
RM
,
Bjorkholm
M
,
Blimark
C
,
Mellqvist
UH
, et al
Monoclonal gammopathy of undetermined significance and risk of skeletal fractures: a population-based study
.
Blood
2010
;
116
:
2651
5
.
44.
Tian
E
,
Zhan
F
,
Walker
R
,
Rasmussen
E
,
Ma
Y
,
Barlogie
B
, et al
The role of the Wnt-signaling antagonist DKK1 in the development of osteolytic lesions in multiple myeloma
.
N Engl J Med
2003
;
349
:
2483
94
.
45.
Paiva
B
,
Perez-Andres
M
,
Vidriales
MB
,
Almeida
J
,
de las Heras
N
,
Mateos
MV
, et al
Competition between clonal plasma cells and normal cells for potentially overlapping bone marrow niches is associated with a progressively altered cellular distribution in MGUS vs myeloma
.
Leukemia
2011
;
25
:
697
706
.
46.
Perez-Andres
M
,
Almeida
J
,
Martin-Ayuso
M
,
Moro
MJ
,
Martin-Nunez
G
,
Galende
J
, et al
Characterization of bone marrow T cells in monoclonal gammopathy of undetermined significance, multiple myeloma, and plasma cell leukemia demonstrates increased infiltration by cytotoxic/Th1 T cells demonstrating a squed TCR-Vbeta repertoire
.
Cancer
2006
;
106
:
1296
305
.
47.
Halapi
E
,
Werner
A
,
Wahlstrom
J
,
Osterborg
A
,
Jeddi-Tehrani
M
,
Yi
Q
, et al
T cell repertoire in patients with multiple myeloma and monoclonal gammopathy of undetermined significance: clonal CD8+ T cell expansions are found preferentially in patients with a low tumor burden
.
Eur J Immunol
1997
;
27
:
2245
52
.
48.
Dhodapkar
MV
,
Krasovsky
J
,
Osman
K
,
Geller
MD
. 
Vigorous premalignancy-specific effector T cell response in the bone marrow of patients with monoclonal gammopathy
.
J Exp Med
2003
;
198
:
1753
7
.
49.
Dhodapkar
MV
,
Geller
MD
,
Chang
DH
,
Shimizu
K
,
Fujii
S
,
Dhodapkar
KM
, et al
A reversible defect in natural killer T cell function characterizes the progression of premalignant to malignant multiple myeloma
.
J Exp Med
2003
;
197
:
1667
76
.
50.
Mahtouk
K
,
Moreaux
J
,
Hose
D
,
Reme
T
,
Meissner
T
,
Jourdan
M
, et al
Growth factors in multiple myeloma: a comprehensive analysis of their expression in tumor cells and bone marrow environment using Affymetrix microarrays
.
BMC Cancer
2010
;
10
:
198
.
51.
Podar
K
,
Richardson
PG
,
Hideshima
T
,
Chauhan
D
,
Anderson
KC
. 
The malignant clone and the bone-marrow environment
.
Best Pract Res Clin Haematol
2007
;
20
:
597
612
.
52.
Kyle
RA
,
Therneau
TM
,
Rajkumar
SV
,
Offord
JR
,
Larson
DR
,
Plevak
MF
, et al
A long-term study of prognosis in monoclonal gammopathy of undetermined significance
.
N Engl J Med
2002
;
346
:
564
9
.
53.
Rosinol
L
,
Cibeira
MT
,
Montoto
S
,
Rozman
M
,
Esteve
J
,
Filella
X
, et al
Monoclonal gammopathy of undetermined significance: predictors of malignant transformation and recognition of an evolving type characterized by a progressive increase in M protein size
.
Mayo Clin Proc
2007
;
82
:
428
34
.
54.
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
.
55.
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
.
56.
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
.
57.
Rajkumar
SV
,
Larson
D
,
Kyle
RA
. 
Diagnosis of smoldering multiple myeloma
.
N Engl J Med
2011
;
365
:
474
5
.
58.
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
.
59.
Cesana
C
,
Klersy
C
,
Barbarano
L
,
Nosari
AM
,
Crugnola
M
,
Pungolino
E
, et al
Prognostic factors for malignant transformation in monoclonal gammopathy of undetermined significance and smoldering multiple myeloma
.
J Clin Oncol
2002
;
20
:
1625
34
.
60.
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
.
61.
Wang
M
,
Alexanian
R
,
Delasalle
K
,
Weber
D
. 
Abnormal MRI of spine is dominant risk factor for early progression of asymptomatic multiple myeloma
.
Blood
2003
;
102
:
687a
.
62.
Katzmann
JA
,
Clark
R
,
Kyle
RA
,
Larson
DR
,
Therneau
TM
,
Melton
LJ
 III
, et al
Suppression of uninvolved immunoglobulins defined by heavy/light chain pair suppression is a risk factor for progression of MGUS
.
Leukemia
.
Epub 2012 Jul 11
.
63.
Kumar
S
,
Rajkumar
SV
,
Kyle
RA
,
Lacy
MQ
,
Dispenzieri
A
,
Fonseca
R
, et al
Prognostic value of circulating plasma cells in monoclonal gammopathy of undetermined significance
.
J Clin Oncol
2005
;
23
:
5668
74
.
64.
La Raja
M
,
Barcobello
M
,
Bet
N
,
Dolfini
P
,
Florean
M
,
Tomasella
F
, et al
Incidental finding of monoclonal gammopathy in blood donors: a follow-up study
.
Blood Transfus
2012
;
10
:
338
43
.
65.
Malacrida
V
,
De Francesco
D
,
Banfi
G
,
Porta
FA
,
Riches
PG
. 
Laboratory investigation of monoclonal gammopathy during 10 years of screening in a general hospital
.
J Clin Pathol
1987
;
40
:
793
7
.
66.
Axelsson
U
,
Bachmann
R
,
Hallen
J
. 
Frequency of pathological proteins (M-components) om 6,995 sera from an adult population
.
Acta Med Scand
1966
;
179
:
235
47
.
67.
Hjorth
M
,
Hellquist
L
,
Holmberg
E
,
Magnusson
B
,
Rodjer
S
,
Westin
J
. 
Initial versus deferred melphalan-prednisone therapy for asymptomatic multiple myeloma stage I–a randomized study. Myeloma Group of Western Sweden
.
Eur J Haematol
1993
;
50
:
95
102
.
68.
Rajkumar
SV
,
Dispenzieri
A
,
Fonseca
R
,
Lacy
MQ
,
Geyer
S
,
Lust
JA
, et al
Thalidomide for previously untreated indolent or smoldering multiple myeloma
.
Leukemia
2001
;
15
:
1274
6
.
69.
Barlogie
B
,
van Rhee
F
,
Shaughnessy
JD
 Jr
,
Epstein
J
,
Yaccoby
S
,
Pineda-Roman
M
, et al
Seven-year median time to progression with thalidomide for smoldering myeloma: partial response identifies subset requiring earlier salvage therapy for symptomatic disease
.
Blood
2008
;
112
:
3122
5
.
70.
Musto
P
,
Petrucci
MT
,
Bringhen
S
,
Guglielmelli
T
,
Caravita
T
,
Bongarzoni
V
, et al
A multicenter, randomized clinical trial comparing zoledronic acid versus observation in patients with asymptomatic myeloma
.
Cancer
2008
;
113
:
1588
95
.
71.
Golombick
T
,
Diamond
TH
,
Manoharan
A
,
Ramakrishna
R
. 
Monoclonal gammopathy of undetermined significance, smoldering multiple myeloma, and curcumin: a randomized, double-blind placebo-controlled cross-over 4g study and an open-label 8g extension study
.
Am J Hematol
2012
;
87
:
455
60
.
72.
Mateos
M
,
López-Corral
L
,
Hernandez
M
,
Giraldo
P
,
Rubia
JDL
,
Arriba
Fd
, 
Smoldering multiple myeloma (SMM) at high-risk of progression to symptomatic disease: a phase III, randomized, multicenter trial based on lenalidomide-dexamethasone (Len-Dex) as induction therapy followed by maintenance therapy with Len alone vs no treatment [abstract]
. In:
Proceedings of the 53rd ASH Annual Meeting and Exposition; 2011 Dec 10–13; San Diego, CA
.
Washington, D.C.
:
ASH
; 
2011
.
Abstract nr 991
.
73.
ClinicalTrials.Gov
.
[Accessed 2012 Nov 4]. Available from
: www.clinicaltrials.gov.