Download as pdf or txt
Download as pdf or txt
You are on page 1of 8

Clinical Review & Education

JAMA Oncology | Review

Moving From Cancer Burden to Cancer Genomics


for Smoldering Myeloma
A Review
Francesco Maura, MD; Niccolò Bolli, MD, PhD; Even H. Rustad, MD, PhD; Malin Hultcrantz, MD, PhD;
Nikhil Munshi, MD; Ola Landgren, MD, PhD

Supplemental content
IMPORTANCE All patients who develop multiple myeloma have a preceding asymptomatic
expansion of clonal plasma cells, clinically recognized as monoclonal gammopathy of
undetermined significance or smoldering multiple myeloma (SMM). During the past decade,
significant progress has been made in the classification and risk stratification of SMM.

OBSERVATIONS This review summarizes current clinical challenges and discusses available
models for risk stratification in the context of SMM. Owing to several novel, more effective,
and less toxic drugs, these aspects are becoming increasingly important to identify patients
eligible for early treatment. However, all proposed criteria were built around indirect markers
of disease burden and therefore are generally able to identify a fraction of patients with SMM
in whom transformation to multiple myeloma and genomic subclonal diversification are
already happening. In contrast, next-generation sequencing approaches have the potential to
identify myeloma precursor disease that will progress even before the major clonal expansion
and progression, providing a potential base for more effective treatment and better precision
regarding the optimal timing of treatment initiation. Author Affiliations: Author
affiliations are listed at the end of this
CONCLUSIONS AND RELEVANCE Owing to modern technologies, in the near future, prognostic article.
models derived from genomic signatures independent of the disease burden will allow better Corresponding Authors: Ola
identification of the optimal timing to treat a plasma cell clonal disorder at the very early Landgren, MD, PhD (landgrec@
stages, when the chances of eradication are higher. mskcc.org), and Francesco Maura,
MD (mauraf@mskcc.org), Myeloma
Service, Department of Medicine,
JAMA Oncol. doi:10.1001/jamaoncol.2019.4659 Memorial Sloan Kettering Cancer
Published online December 12, 2019. Center, 1275 York Ave, New York, NY
10065.

M
any cancers develop through a Darwinian evolution with myeloma precursors who might benefit from early interven-
model along preferred evolutionary trajectories in which tion. In this article, we review the current criteria for risk stratifica-
different driver events accumulate over time and con- tion in SMM and highlight new research and clinical perspectives in
fer a proliferation and survival advantage to a given subclone, pro- light of novel discoveries in the era of genomics.
gressively shaping the genomic profile and the clinical behavior of
that cancer.1 One of the fields in which the concept of premalignant
clonal evolution was described for the first time is gammopathies.
Diagnosis and Assessment
Almost all patients who develop multiple myeloma have a preced-
ing asymptomatic expansion of clonal plasma cells, clinically recog- In an investigation of a retrospective cohort of 276 patients with
nized as monoclonal gammopathy of undetermined significance SMM, Kyle et al10 reported an overall risk of progression to multiple
(MGUS) or smoldering myeloma (SMM).2-6 For more than 20 years, myeloma of 10% per year for the first 5 years, approximately 3% per
the definition and differentiation between these 2 clonal entities year for the next 5 years, and 1% per year for the last 10 years
have been based on indirect measurements of the clonal burden (Figure 1A). That study was the first to report that different serum
(Box).3,4,7,8 These precursor conditions (ie, MGUS and SMM) pro- markers were associated with the risk of SMM progression to mul-
vide an extraordinary opportunity to study the multistep evolution tiple myeloma. The ability to estimate the risk of progression was a
of cancer and to design preventive and early treatment strategies. turning point in the study of SMM, stimulating many different groups
In the past 10 years, the introduction of new drugs has substan- to develop translational studies and clinical trials.
tially changed and improved the treatment of multiple myeloma After these preliminary observations, different markers were
(eFigure 1 in the Supplement).9 Many of these new drugs are more subsequently reported to be associated with an increased risk of
effective and less toxic than traditional chemotherapy, providing for SMM progression, and 2 major models for SMM risk stratification
the first time attractive agents for early treatment strategies. By com- were developed based on combinations of these markers (Figure 1B
bining these novel regimens with modern technologies, such as next- and eTable 1 in the Supplement)12-16: (1) the Mayo Clinic model, mostly
generation sequencing, we have the opportunity to identify patients focused on serum protein abnormalities as surrogate measures of

jamaoncology.com (Reprinted) JAMA Oncology Published online December 12, 2019 E1

© 2019 American Medical Association. All rights reserved.

Downloaded From: https://jamanetwork.com/ by a Western University User on 12/12/2019


Clinical Review & Education Review Moving From Cancer Burden to Cancer Genomics for Smoldering Myeloma

tumor burden,10 and (2) the Programa para el Tratamiento de


Hemopatias Malignas (PETHEMA) model, using multiparametric flow Box. Definition of Multiple Myeloma Based on 2014 International
cytometry to phenotype bone marrow plasma cells.17 A head-to- Myeloma Working Group Criteria
head comparison11 of the Mayo Clinic and PETHEMA models in a pro-
spective cohort of 77 patients with SMM found a significant discor- Criteria for Multiple Myeloma
Clonal bone marrow plasma cells of at least 10% or biopsy-proven
dance in the overall risk stratification, with only 28.6% of patients
bony or extramedullary plasmacytoma and any 1 or more of the
overlapping on specific risk categories: low risk vs high risk, low risk following myeloma-defining events:
vs non–low risk, and high risk vs non–high risk (Figure 1C). Further- 1. Evidence of end-organ damage that can be attributed to the
more, in the Swedish Myeloma Registry, only 30% of SMM cases that underlying plasma cell proliferative disorder, specifically:
progressed met the Mayo Clinic criteria for high-risk disease.18 Over- • Hypercalcemia: serum calcium level greater than 1 mg/dL
all, this finding highlights the need for more robust strategies to iden- higher than the upper limit of normal or greater than 11 mg/dL
(to convert to micromoles per liter, multiply by 0.25)
tify individuals with myeloma precursor disease who are at high risk
• Renal insufficiency: creatinine clearance <0.67 mL/s/m2
of progression.
(to convert to milliliters per minute per 1.73 m2, multiply by
Owing to technological advances, several different ap- 0.0167) or serum creatinine level greater than 2 mg/dL
proaches have been tested to allow an earlier and more accurate (to convert to micromoles per liter, multiply by 88.4)
identification of patients with high-risk SMM. The application of • Anemia: hemoglobin value greater than 2.0 g/dL below the
imaging approaches, such as magnetic resonance imaging of the lower limit of normal or a hemoglobin value less than 10.0
spine and pelvis and whole-body positron emission tomography– g/dL (to convert to grams per liter, multiply by 10)
• Bone lesions: 1 or more osteolytic lesions on skeletal
computed tomography, were used to estimating the risk of progres-
radiography, computed tomography, or positron emission
sion to multiple myeloma.19-21 Specifically, patients with focal le-
tomography–computed tomography
sions and/or bone marrow abnormalities were characterized by 2. Any 1 or more of the following biomarkers of malignancy:
a higher risk of progression.22 However, these approaches still re- • Clonal bone marrow plasma cell percentage of 60% or
flect the SMM disease burden and end-organ damage. greater
Of importance, alternative efforts have been attempted to • Involved-uninvolved serum free light-chain ratio of 100 or
identify markers of SMM progression that are not measures of the greater
• More than 1 focal lesion on magnetic resonance imaging
disease burden but rather are more intrinsically linked to the under-
lying biological features of the disease. In the past 20 years, several Definition of Smoldering Multiple Myeloma
studies23,24 reported recurrent translocations and copy number 1. Serum monoclonal protein (IgG or IgA) level of 3000 mg/dL or
abnormalities (CNAs) in patients with multiple myeloma using greater (to convert to grams per liter, multiply by 0.01) or
urinary monoclonal protein level of 500 mg/24 hours or
karyotype and fluorescence in situ hybridization, with some of these
greater and/or clonal bone marrow plasma cell count of 10% to
translocations and CNAs having associations with prognosis 60%
(Figure 2). On the basis of these efforts, recurrent CNAs and trans- 2. Absence of myeloma-defining events or amyloidosis
locations were investigated by fluorescence in situ hybridization in
several different clinical trials and retrospective cohort studies, which
showed that the presence of del17p13 (TP53), t(4;14)(MMSET;IGH), genomic background more prone to acquire new lesions and mi-
and/or 1q21 gain were associated with shortened time to multiple croenvironment changes with the potential to lead to an aggressive
myeloma progression in SMM and MGUS.30,31 Using a different ap- evolutionary trajectory and consequently to a shift in clinical behav-
proach, Dhodapkar et al32 identified a 70-gene expression signa- ior. In contrast to the first group, the key driver events associated with
ture in SMM (GEP70) that was independently associated with the progression to multiple myeloma have not yet been acquired in this
risk of progression to multiple myeloma. However, similar to many intermediate group. The third group is characterized by a clinical
prognostic models based on gene expression, the application of this course compatible with that of MGUS despite meeting clinical crite-
approach into daily clinical practice has been limited by low stan- ria for SMM diagnosis (ie, they have a recognizable burden of dis-
dardization and low reproducibility. ease in terms of plasma cell bone marrow infiltration and serum M-
component). In this last group of patients, the genomic predisposition
to acquire new driver events associated with progression is low, the
clone is stable, and the clinical course is indolent in the long term. Al-
Prognosis though these patterns can be identified by careful scrutiny of pro-
Overall, when progression curves of SMM are considered, 3 main pat- gression curves, to date, we do not have any direct evidence of the
terns can be identified based on the rate of transformation, likely re- biological events causally linked to disease progression, and it is there-
flecting differences in underlying biological characteristics10 (Figure 1). fore currently not possible to assign a patient to a group at the time
The first pattern is a group characterized by ongoing transforma- of SMM diagnosis. However, the identification of biological markers
tion already at the time of SMM diagnosis who will meet transfor- to estimate MGUS and SMM progression before the onset of end-
mation criteria within a few months or years. These patients have organ damage has always been a main goal of multiple myeloma re-
likely already acquired the key driver events associated with pro- search. Thanks to the wide availability of next-generation sequenc-
gression and likely have a genomic and microenvironment land- ing platforms, we now have the means to address this critical task.
scape similar to the one of multiple myeloma. A second group will In general, MGUS and SMM shared some recurrent genomic
experience progression after several years of an indolent and asymp- aberrations with multiple myeloma (Figure 2 and eTable 2 in the
tomatic clinical course. This group of patients with SMM likely has a Supplement); however, others, such as 13q and 17p deletions,

E2 JAMA Oncology Published online December 12, 2019 (Reprinted) jamaoncology.com

© 2019 American Medical Association. All rights reserved.

Downloaded From: https://jamanetwork.com/ by a Western University User on 12/12/2019


Moving From Cancer Burden to Cancer Genomics for Smoldering Myeloma Review Clinical Review & Education

Figure 1. Clinical Outcome of Multiple Myeloma Precursors

A Evolution trajectories

100
1 2 Established ongoing transformation
3
and ongoing clinical progression
High predisposition to genomic
80
Probability of Progression, %

transformation
SMM Low predisposition to genomic
transformation
60

40
4

20
MGUS

0
0 5 10 15 20 25
C Risk of SMM
Years
1.0
PETHEMA
B Myeloma progression risk
Low
0.8 Intermediate
PETHEMA
High
Low
0.6

Proportion
Intermediate

High
0.4
Mayo
Low
0.2
Intermediate

High
0
0 20 40 60 80 100 Mayo Mayo Mayo
5-y SMM Progression, % Low Intermediate High

A, Three evolution trajectories for monoclonal gammopathy of undetermined by low predisposition to acquire new driver events and progress over the time,
significance (MGUS) and smoldering multiple myeloma (SMM) progression with a clinical outcome similar to MGUS. B, Risk of SMM or MGUS progression
(adapted from Kyle et al10), potentially reflecting 3 different biological and according to the Mayo Clinic model or Programa para el Tratamiento de
genomic profiles. The first trajectories reflect an ongoing transformation and Hemopatias Malignas (PETHEMA) prognostic model. C, Comparison between
progression. The second reflect patients and clones with high predisposition to the Mayo Clinic and PETHEMA risk models for the identification of patients with
acquire new driver events and progress over the time. The third is characterized high-risk SMM. Adapted from Cherry et al.11

were observed at lower prevalence compared with multiple the MGUS and SMM genomic landscape is less complex than that
myeloma.25-29,31,33,34 Furthermore, all but t(11;14)(CCND1;IGH) of multiple myeloma and the identification of secondary events in-
recurrent cytogenetic and structural aberrations have been volved in progression could represent an important early and accu-
reported at lower frequency among MGUS cases compared with rate prognostic marker. However, such identification is not an easy
multiple myeloma and SMM cases (Figure 2 and eTable 2 in the task in multiple myeloma. Some hematologic malignant tumors, such
Supplement). This finding confirms the existence of a chronologic as acute myeloid leukemia, are usually characterized by a low ge-
order of acquisition of cytogenetic aberrations potentially nomic complexity, and the characterization of their genomic land-
involved in the clone progression from MGUS to SMM and from scape based on targeted and/or exome-based approaches has been
SMM to multiple myeloma.35 relatively straightforward.47 Similar approaches have been at-
Using whole genome sequencing and whole exome sequenc- tempted in multiple myeloma40,43,48,49; however, its genomic land-
ing data, different groups have recently found that multiple my- scape is more complex and heterogeneous (Figure 3C).42 The first
eloma has a complex and heterogeneous genomic profile.36-41 Sig- whole genome sequencing characterization of 10 non–high-risk
nificant efforts have been made to characterize the landscape of SMM cases that progressed to multiple myeloma highlighted the
driver mutations in multiple myeloma genes.36,37,39,42,43 However, great genomic complexity and heterogeneity of this premalignant
several reports39,42,43 suggest that driver mutations are secondary entity.44 Two different progression models were reported based on
events and not directly involved in the cancer initiation and early pro- gene mutations, translocations, CNAs, and mutational signatures in
gression. To confirm this theory, initial whole exome sequencing and paired SMM and multiple myeloma samples (eFigure 2 in the Supple-
whole genome sequencing studies28,44-46 have found a lower preva- ment): (1) the static model, in which the same subclonal architec-
lence of these driver mutations in MGUS and SMM compared with ture was retained as the disease progressed to multiple myeloma,
what has been reported among newly diagnosed multiple my- and (2) the spontaneous evolution model, in which the subclonal
eloma (Figure 3A and B). Overall, these observations suggest that composition of SMM changed without any external selective

jamaoncology.com (Reprinted) JAMA Oncology Published online December 12, 2019 E3

© 2019 American Medical Association. All rights reserved.

Downloaded From: https://jamanetwork.com/ by a Western University User on 12/12/2019


Clinical Review & Education Review Moving From Cancer Burden to Cancer Genomics for Smoldering Myeloma

onstration of the role of the microenvironment in progression of


Figure 2. Multiple Myeloma (MM) Cytogenetic Precursor
monoclonal gammopathies came from the study of a humanized
0.6 mouse model,58 in which xenografts from patients with asymptom-
MGUS SMM MM atic gammopathies showed progressive growth, suggesting that the
0.5
clinical stability of these conditions may be in part caused by
extrinsic growth restriction of tumor cells from the host microen-
Proportion of Patients

0.4
vironment. Of interest, progression occurred after a Darwinian
0.3
competition during which a minor subclone in the patient
acquired clonal dominance in the xenograft. This finding is an
0.2 example of how the heterogeneous genomic landscape of plasma
cell disorders can dynamically adapt to different microenviron-
0.1 ments in an interplay required for tumor progression. This con-
cept was also supported by a study59 on simultaneous biopsies in
0
t(11;14) t(4;14) t(14;16) HRD 13q del 17p del 1q gain newly diagnosed multiple myeloma that revealed an extensive
or t(14;14) heterogeneity among spatially distinct lesions, possibly reflecting
the influence of different microenvironmental contexts on sub-
The prevalence of the main cytogenetic aberration evaluated by fluorescence
clonal selection within tumor cells.
in situ hybridization or single-nucleotide polymorphism array among different
published cohorts of monoclonal gammopathy of undetermined significance Recently, the first large, population-based prospective screen-
(MGUS), smoldering multiple myeloma (SMM), and newly diagnosed MMs.25-29 ing study60 from myeloma precursor disease to multiple myeloma
provided novel insight on how serum immune markers—indicators
of disease burden—can change over time, emphasizing the impor-
pressure. Although patients included in the first group generally tance of repeated blood tests and reassessment of risk annually. A
experienced progression in less than 1 year, reflecting an already logical extension of the results from the population-based prospec-
ongoing transformation process, the second had a longer time to tive screening study60 is the application of genomic assays on lon-
progression. Although the identification of robust novel prog- gitudinally collected samples, with the aim of defining key evolu-
nostic markers of SMM progression was beyond the scope of the tionary and clinical trajectories involved in the progression from
study, additional studies 42,44,46,50-52 confirmed that several myeloma precursor disease to multiple myeloma.
genomic lesions only explorable by whole genome sequencing The integration of cell-intrinsic and cell-extrinsic analyses will
might be involved in SMM progression. Aside from mutations, offer the opportunity to design new prognostic models that are as-
CNAs, and translocations, analysis of mutational signatures can sociated with biological features of the disease rather than with its
also provide clues to disease progression. In particular, aberrant burden. Harnessing the selective evolutionary pressure from the
activity of the APOBEC family of DNA deaminases, a mutational microenvironment for therapeutic purposes, before a more active
process particularly active in aggressive multiple myeloma,41,50 subclone takes over and the disease (eventually) becomes symp-
was mostly present in late SMM subclones, suggesting a role in tomatic, will also be a relevant challenge for the future.
progression to multiple myeloma.
The ability to identify biological hallmarks of disease aggres- Treatment
siveness among patients with MGUS and SMM has been further con- The current standard of care for patients with multiple myeloma pre-
firmed by a recent single-cell RNA sequencing study53 in which the cursors is watchful waiting. The rationale for this approach comes
authors found small clonal populations in patients with MGUS that from unsuccessful early treatment approaches using melphalan-
were indistinguishable at the molecular level from those in individu- prednisone, thalidomide, or bisphosphonates.61-67
als with multiple myeloma. Because of the increased access to novel drugs that are highly
One limitation of genomic studies is that they are focused on effective and generally safe, early high-risk SMM treatment is in the
the cancer (ie, myeloma) cells and thus blind to the complex inter- spotlight again. Rationale for early treatment of patients with SMM
action with microenvironment. The cancer-naive niche is not suited consists of better potential of disease control, if not cure, in the early
for cancer cell growth. Cancer cells may, however, acquire the abil- stages in which the subclonal complexity (and hence drug resis-
ity to induce phenotypic changes in the microenvironment, pro- tance) could be lower. Furthermore, patients may better tolerate
moting the production of cellular and humoral factors that may sus- treatment in the absence of end-organ damage. However, there is
tain cancer proliferation, as well as escape from immune tumor a risk of overtreating a fraction of patients who would otherwise be
surveillance to make it permissive to cancer cell seeding.54,55 Over- stable for years. In addition, treatment could represent an early
all, the microenvironment can influence the myeloma genesis in dif- bottleneck of evolution for cancer cells, thereby promoting earlier
ferent ways: (1) by creating the condition for the cancer initiations expansion of more aggressive and refractory clones compared with
within the germinal center, (2) by inducing a selective pressure lim- conventional watch-and-wait approaches. Furthermore, although
iting the expansion of premalignant entities, and (3) by promoting the antimyeloma activity of chemotherapeutical agents is widely
the premalignant clonal expansion and progression into sympto- accepted, their short- and long-term toxic effects would still be con-
matic disease. The role of the microenvironment in multiple my- cerning in the context of SMM.
eloma has been extensively studied, and comparative studies56,57 The 2014 updated diagnostic criteria for multiple myeloma sug-
have highlighted changes that may be associated with progression gested that ultra–high-risk SMM (now defined as multiple my-
from asymptomatic and symptomatic stages. A direct, in vivo dem- eloma) should be treated even if asymptomatic, owing to a 80% risk

E4 JAMA Oncology Published online December 12, 2019 (Reprinted) jamaoncology.com

© 2019 American Medical Association. All rights reserved.

Downloaded From: https://jamanetwork.com/ by a Western University User on 12/12/2019


Moving From Cancer Burden to Cancer Genomics for Smoldering Myeloma Review Clinical Review & Education

Figure 3. Genomic Landscape of Multiple Myeloma (MM) Precursors

A Next-generation sequencing studies B CoMMpass AI11

Mailankody Bolli Walker


et al45 et al44 et al46 Mikulasova et al27,28
SMM SMM SMM/MGUS MGUS

ACTG1 ACTG1
BRAF BRAF
CYLD CYLD
DIS3 DIS3
FAM46C FAM46C
HIST1H1E HIST1H1E
IRF4 IRF4
KRAS KRAS
LTB LTB
MAX MAX
NRAS NRAS
PRDM1 PRDM1
RB1 RB1
TP53 TP53
TRAF3 TRAF3

C PD26400a

Y 1
X

22
21
20 2

19

18

17 3

16

15 4

14

13

12 6

11
7

10
9 8

A-B, Heatmap showing the prevalence of recurrent MM mutations among SMM whole genome landscape. All main genomic events are reported in this
monoclonal gammopathy of undetermined significance (MGUS), smoldering genome plot: mutations (external circle), indels (middle circle; dark green and
multiple myeloma (SMM), and MMs according to the 4 main next-generation brown lines represent insertion and deletion respectively), copy number
sequencing studies and among newly diagnosed MM enrolled within the variants (red indicates deletions, green indicates gain) and rearrangements
CoMMpass (Relating Clinical Outcomes in MM to Personal Assessment of (blue indicates inversion, red indicates deletions, green indicates tandem
Genetic Profile) trial.28,44-46 The CoMMpass data were generated as part of the duplication, black indicates translocation). This patient had non–high-risk
Multiple Myeloma Research Foundation Personalized Medicine Initiatives SMM10 that progressed after 42 months.
(https://research.themmrf.org and http://www.themmrf.org). C, Example of

jamaoncology.com (Reprinted) JAMA Oncology Published online December 12, 2019 E5

© 2019 American Medical Association. All rights reserved.

Downloaded From: https://jamanetwork.com/ by a Western University User on 12/12/2019


Clinical Review & Education Review Moving From Cancer Burden to Cancer Genomics for Smoldering Myeloma

of developing end-organ damage in the subsequent 2 years.22 For


Figure 4. The Rationale of Early Treatment in Patients With Smoldering
patients with SMM who have a high risk of progression, who do not Multiple Myeloma (SMM) and Monoclonal Gammopathy of
fit the new criteria for multiple myeloma, several attempts have Undetermined Significance (MGUS)
been made to challenge this practice since the advent of novel
drugs. In 2013, Mateos et al68,69 published, to our knowledge, the A Step 1

first phase 3 trial, in which patients with high-risk SMM were ran- Early Late Multiple First
Initiation expansion expansion myeloma Treatment relapse
domized between the watch-and-wait approach and treatment
with lenalidomide-dexamethasone for 9 cycles, followed by a fixed
2-year maintenance regimen. The authors reported a survival
advantage in the experimental arm of 94% vs 80% for patients
alive at 3 years.
In the early 2010s,45,70 studies of the combination of carfil-
B Step 2
zomib, lenalidomide, and dexamethasone, the first 3-drug combi-
Early Late First
nation therapy targeting patients at high risk of multiple myeloma, Initiation expansion expansion Treatment relapse
showed a high rate of minimum residual disease negativity (92%).
Despite results from these trials, follow-up is still short, and the
paradigm of SMM treatment has not changed in routine clinical
practice; however, several clinical trials targeting patients with
high-risk SMM are currently ongoing, and considering the efficacy
of novel agent combinations in newly diagnosed patients, most of C Step 3
these have been designed without standard chemotherapy Early
Initiation expansion Treatment Remission or eradication
(eTable 3 in the Supplement).
Before treatment of SMM can be accepted as standard prac-
tice, several biological and clinical questions must be addressed. Clini-
cal and laboratory markers associated with risk of progression are
mainly based on surrogate measures of disease burden, and the cho-
sen cutoffs are generally based on arbitrary values extracted from Multiple myeloma pathogenesis is characterized by a typical Darwinian
retrospective series; it is a common experience of practitioners that evolution model characterized by accumulation over time of different driver
events along preferred evolutionary trajectories. A, When multiple myeloma is
these risk scores are inaccurate for some patients. We believe that treated, most patients achieve a good response and durable remission.
through genomic studies, we will, in the future, detect aggressive However, a significant fraction still experience progression, generally after a
disease cases in asymptomatic phases, also allowing for control of clonal or subclonal competition and selection induced by treatment. This clonal
heterogeneity and genomic complexity represent the main reasons why it is
subclonal diversification and long-term toxic effects on the hema-
difficult to completely eradicate this cancer. B, A significant fraction of high-risk
topoietic stem cells from treatment (Figure 4). SMM already has the key driver events and has already expanded its clonal and
subclonal architecture, reducing chance to eradicate the disease. C, Conversely,
at the very early stage of MGUS and SMM clonal expansion, the genomic and
clonal complexity is likely lower, potentially increasing the premalignant clone
Conclusions eradication chances.

In the coming years, we need to better understand the biological de-


terminants and the dynamics of evolution of myeloma precursor dis- the current genomic era and will potentially allow practitioners to bet-
ease to identify more robust genomic prognostic markers to be ter identify the optimal timing to treat a plasma cell clonal disorder at
combined within current risk models. This goal is finally achievable in the very early stages, when the chances of eradication are higher.

ARTICLE INFORMATION Author Contributions: Dr Maura had full access to Research Council outside the submitted work.
Accepted for Publication: August 8, 2019. all the data in the study and takes responsibility for Dr Munshi reported receiving grants from Celgene,
the integrity of the data and the accuracy of the receiving personal fees from Oncopep outside the
Published Online: December 12, 2019. data analysis. submitted work, and having a patent to Oncopep
doi:10.1001/jamaoncol.2019.4659 Concept and design: Maura, Bolli, Munshi, issued and licensed. Dr Landgren reported
Author Affiliations: Myeloma Service, Department Landgren. receiving grants and personal fees from Amgen,
of Medicine, Memorial Sloan Kettering Cancer Acquisition, analysis, or interpretation of data: Janssen, Celgene, and Karyopharm; receiving
Center, New York, New York (Maura, Rustad, All authors. personal fees from Adaptive Biotech; and serving
Hultcrantz, Landgren); Cancer Genome Project, Drafting of the manuscript: Maura, Bolli, Landgren. on independent data monitoring committees for
Wellcome Trust Sanger Institute, Cambridgeshire, Critical revision of the manuscript for important Merck, Theradex, and Takeda outside the
United Kingdom (Maura); Department of Oncology intellectual content: All authors. submitted work. No other disclosures were
and Hemato-Oncology, University of Milan, Milan, Statistical analysis: Maura, Hultcrantz. reported.
Italy (Bolli); Department of Hematology, Obtained funding: Landgren. Funding/Support: This study was funded by core
Fondazione IRCCS Istituto Nazionale dei Tumori, Administrative, technical, or material support: grant P30 CA008748 from Memorial Sloan
Milan, Italy (Bolli); Jerome Lipper Multiple Myeloma Maura, Bolli, Munshi, Landgren. Kettering Cancer Center (Drs Maura and Landgren),
Center, Dana-Farber Cancer Institute, Harvard Supervision: Maura, Bolli, Landgren. the Perelman Family Foundation in collaboration
Medical School, Boston, Massachusetts (Munshi); Conflict of Interest Disclosures: Dr Hultcrantz with the Multiple Myeloma Research Foundation
Veterans Administration Boston Healthcare reported receiving grants from the Multiple (Drs Maura and Landgren), and grant 817997 from
System, West Roxbury, Massachusetts (Munshi). Myeloma Research Foundation and the Swedish the European Research Council under the European

E6 JAMA Oncology Published online December 12, 2019 (Reprinted) jamaoncology.com

© 2019 American Medical Association. All rights reserved.

Downloaded From: https://jamanetwork.com/ by a Western University User on 12/12/2019


Moving From Cancer Burden to Cancer Genomics for Smoldering Myeloma Review Clinical Review & Education

Union’s Horizon 2020 research and innovation 14. Landgren O. Monoclonal gammopathy of significance. Eur J Haematol. 2016;97(6):568-575.
program (Dr Bolli). undetermined significance and smoldering multiple doi:10.1111/ejh.12774
Role of the Funder/Sponsor: The funding sources myeloma: biological insights and early treatment 28. Mikulasova A, Wardell CP, Murison A, et al. The
had no role in the design and conduct of the study; strategies. Hematology Am Soc Hematol Educ spectrum of somatic mutations in monoclonal
collection, management, analysis, and Program. 2013;2013:478-487. doi:10.1182/ gammopathy of undetermined significance
interpretation of the data; preparation, review, or asheducation-2013.1.478 indicates a less complex genomic landscape than
approval of the manuscript; and decision to submit 15. Larsen JT, Kumar SK, Dispenzieri A, Kyle RA, that in multiple myeloma. Haematologica. 2017;102
the manuscript for publication. Katzmann JA, Rajkumar SV. Serum free light chain (9):1617-1625. doi:10.3324/haematol.2017.163766
ratio as a biomarker for high-risk smoldering 29. Schmidt-Hieber M, Gutiérrez ML, Pérez-Andrés
REFERENCES multiple myeloma. Leukemia. 2013;27(4):941-946. M, et al. Cytogenetic profiles in multiple myeloma
1. Stratton MR, Campbell PJ, Futreal PA. The cancer doi:10.1038/leu.2012.296 and monoclonal gammopathy of undetermined
genome. Nature. 2009;458(7239):719-724. doi: 16. Rajkumar SV, Larson D, Kyle RA. Diagnosis of significance: a study in highly purified aberrant
10.1038/nature07943 smoldering multiple myeloma. N Engl J Med. 2011; plasma cells. Haematologica. 2013;98(2):279-287.
2. Landgren O, Weiss BM. Patterns of monoclonal 365(5):474-475. doi:10.1056/NEJMc1106428 doi:10.3324/haematol.2011.060632
gammopathy of undetermined significance and 17. Pérez-Persona E, Vidriales MB, Mateo G, et al. 30. Neben K, Jauch A, Hielscher T, et al.
multiple myeloma in various ethnic/racial groups: New criteria to identify risk of progression in Progression in smoldering myeloma is
support for genetic factors in pathogenesis. monoclonal gammopathy of uncertain significance independently determined by the chromosomal
Leukemia. 2009;23(10):1691-1697. doi:10.1038/leu. and smoldering multiple myeloma based on abnormalities del(17p), t(4;14), gain 1q,
2009.134 multiparameter flow cytometry analysis of bone hyperdiploidy, and tumor load. J Clin Oncol. 2013;31
3. Kyle RA, Therneau TM, Rajkumar SV, et al. marrow plasma cells. Blood. 2007;110(7):2586-2592. (34):4325-4332. doi:10.1200/JCO.2012.48.4923
Prevalence of monoclonal gammopathy of doi:10.1182/blood-2007-05-088443 31. Rajkumar SV, Gupta V, Fonseca R, et al. Impact
undetermined significance. N Engl J Med. 2006; 18. Kristinsson SY, Holmberg E, Blimark C. of primary molecular cytogenetic abnormalities and
354(13):1362-1369. doi:10.1056/NEJMoa054494 Treatment for high-risk smoldering myeloma. risk of progression in smoldering multiple myeloma.
4. Kyle RA, Greipp PR. Smoldering multiple N Engl J Med. 2013;369(18):1762-1763. doi:10.1056/ Leukemia. 2013;27(8):1738-1744. doi:10.1038/leu.
myeloma. N Engl J Med. 1980;302(24):1347-1349. NEJMc1310911 2013.86
doi:10.1056/NEJM198006123022405 19. Hillengass J, Fechtner K, Weber MA, et al. 32. Dhodapkar MV, Sexton R, Waheed S, et al.
5. Fidler IJ. Tumor heterogeneity and the biology of Prognostic significance of focal lesions in Clinical, genomic, and imaging predictors of
cancer invasion and metastasis. Cancer Res. 1978; whole-body magnetic resonance imaging in myeloma progression from asymptomatic
38(9):2651-2660. patients with asymptomatic multiple myeloma. monoclonal gammopathies (SWOG S0120). Blood.
J Clin Oncol. 2010;28(9):1606-1610. doi:10.1200/ 2014;123(1):78-85. doi:10.1182/blood-2013-07-515239
6. Dexter DL, Kowalski HM, Blazar BA, Fligiel Z, JCO.2009.25.5356
Vogel R, Heppner GH. Heterogeneity of tumor cells 33. Brousseau M, Leleu X, Gerard J, et al;
from a single mouse mammary tumor. Cancer Res. 20. Kastritis E, Moulopoulos LA, Terpos E, Intergroupe Francophone du Myélome.
1978;38(10):3174-3181. Koutoulidis V, Dimopoulos MA. The prognostic Hyperdiploidy is a common finding in monoclonal
importance of the presence of more than one focal gammopathy of undetermined significance and
7. Rajkumar SV, Landgren O, Mateos MV. lesion in spine MRI of patients with asymptomatic monosomy 13 is restricted to these hyperdiploid
Smoldering multiple myeloma. Blood. 2015;125(20): (smoldering) multiple myeloma. Leukemia. 2014;28 patients. Clin Cancer Res. 2007;13(20):6026-6031.
3069-3075. doi:10.1182/blood-2014-09-568899 (12):2402-2403. doi:10.1038/leu.2014.230 doi:10.1158/1078-0432.CCR-07-0031
8. Landgren O, Kyle RA, Pfeiffer RM, et al. 21. Zamagni E, Nanni C, Gay F, et al. 18F-FDG 34. Chng WJ, Van Wier SA, Ahmann GJ, et al.
Monoclonal gammopathy of undetermined PET/CT focal, but not osteolytic, lesions predict the A validated FISH trisomy index demonstrates the
significance (MGUS) consistently precedes multiple progression of smoldering myeloma to active hyperdiploid and nonhyperdiploid dichotomy in
myeloma: a prospective study. Blood. 2009;113(22): disease. Leukemia. 2016;30(2):417-422. doi:10. MGUS. Blood. 2005;106(6):2156-2161. doi:10.1182/
5412-5417. doi:10.1182/blood-2008-12-194241 1038/leu.2015.291 blood-2005-02-0761
9. Gay F, Jackson G, Rosiñol L, et al. Maintenance 22. Rajkumar SV, Dimopoulos MA, Palumbo A, 35. van Nieuwenhuijzen N, Spaan I, Raymakers R,
treatment and survival in patients with myeloma: et al. International Myeloma Working Group Peperzak V. From MGUS to multiple myeloma, a
a systematic review and network meta-analysis. updated criteria for the diagnosis of multiple paradigm for clonal evolution of premalignant cells.
JAMA Oncol. 2018;4(10):1389-1397. doi:10.1001/ myeloma. Lancet Oncol. 2014;15(12):e538-e548. Cancer Res. 2018;78(10):2449-2456. doi:10.1158/
jamaoncol.2018.2961 doi:10.1016/S1470-2045(14)70442-5 0008-5472.CAN-17-3115
10. Kyle RA, Remstein ED, Therneau TM, et al. 23. Manier S, Salem KZ, Park J, Landau DA, Getz G, 36. Bolli N, Avet-Loiseau H, Wedge DC, et al.
Clinical course and prognosis of smoldering Ghobrial IM. Genomic complexity of multiple Heterogeneity of genomic evolution and
(asymptomatic) multiple myeloma. N Engl J Med. myeloma and its clinical implications. Nat Rev Clin mutational profiles in multiple myeloma. Nat
2007;356(25):2582-2590. doi:10.1056/ Oncol. 2017;14(2):100-113. doi:10.1038/nrclinonc. Commun. 2014;5:2997. doi:10.1038/ncomms3997
NEJMoa070389 2016.122 37. Chapman MA, Lawrence MS, Keats JJ, et al.
11. Cherry BM, Korde N, Kwok M, et al. Modeling 24. Morgan GJ, Walker BA, Davies FE. The genetic Initial genome sequencing and analysis of multiple
progression risk for smoldering multiple myeloma: architecture of multiple myeloma. Nat Rev Cancer. myeloma. Nature. 2011;471(7339):467-472. doi:10.
results from a prospective clinical study. Leuk 2012;12(5):335-348. doi:10.1038/nrc3257 1038/nature09837
Lymphoma. 2013;54(10):2215-2218. doi:10.3109/
10428194.2013.764419 25. Lakshman A, Rajkumar SV, Buadi FK, et al. Risk 38. Keats JJ, Chesi M, Egan JB, et al. Clonal
stratification of smoldering multiple myeloma competition with alternating dominance in multiple
12. Agarwal A, Ghobrial IM. Monoclonal incorporating revised IMWG diagnostic criteria. myeloma. Blood. 2012;120(5):1067-1076. doi:10.
gammopathy of undetermined significance and Blood Cancer J. 2018;8(6):59. doi:10.1038/s41408- 1182/blood-2012-01-405985
smoldering multiple myeloma: a review of the 018-0077-4
current understanding of epidemiology, biology, 39. Lohr JG, Stojanov P, Carter SL, et al; Multiple
risk stratification, and management of myeloma 26. López-Corral L, Gutiérrez NC, Vidriales MB, Myeloma Research Consortium. Widespread
precursor disease. Clin Cancer Res. 2013;19(5):985- et al. The progression from MGUS to smoldering genetic heterogeneity in multiple myeloma:
994. doi:10.1158/1078-0432.CCR-12-2922 myeloma and eventually to multiple myeloma implications for targeted therapy. Cancer Cell. 2014;
involves a clonal expansion of genetically abnormal 25(1):91-101. doi:10.1016/j.ccr.2013.12.015
13. Dispenzieri A, Stewart AK, Chanan-Khan A, plasma cells. Clin Cancer Res. 2011;17(7):1692-1700.
et al. Smoldering multiple myeloma requiring 40. Walker BA, Boyle EM, Wardell CP, et al.
doi:10.1158/1078-0432.CCR-10-1066 Mutational spectrum, copy number changes, and
treatment: time for a new definition? Blood. 2013;
122(26):4172-4181. doi:10.1182/blood-2013-08- 27. Mikulasova A, Smetana J, Wayhelova M, et al. outcome: results of a sequencing study of patients
520890 Genomewide profiling of copy-number alteration in with newly diagnosed myeloma. J Clin Oncol. 2015;
monoclonal gammopathy of undetermined 33(33):3911-3920. doi:10.1200/JCO.2014.59.1503

jamaoncology.com (Reprinted) JAMA Oncology Published online December 12, 2019 E7

© 2019 American Medical Association. All rights reserved.

Downloaded From: https://jamanetwork.com/ by a Western University User on 12/12/2019


Clinical Review & Education Review Moving From Cancer Burden to Cancer Genomics for Smoldering Myeloma

41. Walker BA, Wardell CP, Murison A, et al. 52. Dutta AK, Fink JL, Grady JP, et al. Subclonal prednisone therapy for asymptomatic multiple
APOBEC family mutational signatures are evolution in disease progression from MGUS/SMM myeloma stage I: a randomized study. Eur J Haematol.
associated with poor prognosis translocations in to multiple myeloma is characterised by clonal 1993;50(2):95-102. doi:10.1111/j.1600-0609.1993.
multiple myeloma. Nat Commun. 2015;6:6997. doi: stability. Leukemia. 2019;33(2):457-468. doi:10. tb00148.x
10.1038/ncomms7997 1038/s41375-018-0206-x 63. Martín A, García-Sanz R, Hernández J, et al.
42. Maura F, Bolli N, Angelopoulos N, et al. 53. Ledergor G, Weiner A, Zada M, et al. Single cell Pamidronate induces bone formation in patients with
Genomic landscape and chronological dissection of plasma cell heterogeneity in smouldering or indolent myeloma, with no significant
reconstruction of driver events in multiple symptomatic and asymptomatic myeloma. Nat Med. anti-tumour effect. Br J Haematol. 2002;118(1):
myeloma. Nat Commun. 2019;10(1):3835. doi:10. 2018;24(12):1867-1876. doi:10.1038/s41591-018- 239-242. doi:10.1046/j.1365-2141.2002.03549.x
1038/s41467-019-11680-1 0269-2 64. Musto P, Petrucci MT, Bringhen S, et al;
43. Walker BA, Mavrommatis K, Wardell CP, et al. 54. Valastyan S, Weinberg RA. Tumor metastasis: GIMEMA (Italian Group for Adult Hematologic
Identification of novel mutational drivers reveals molecular insights and evolving paradigms. Cell. Diseases)/Multiple Myeloma Working Party and the
oncogene dependencies in multiple myeloma. Blood. 2011;147(2):275-292. doi:10.1016/j.cell.2011.09.024 Italian Myeloma Network. A multicenter,
2018;132(6):587-597. doi:10.1182/blood-2018-03- 55. Fidler IJ. The pathogenesis of cancer randomized clinical trial comparing zoledronic acid
840132 metastasis: the ‘seed and soil’ hypothesis revisited. versus observation in patients with asymptomatic
44. Bolli N, Maura F, Minvielle S, et al. Genomic Nat Rev Cancer. 2003;3(6):453-458. doi:10.1038/ myeloma. Cancer. 2008;113(7):1588-1595. doi:10.
patterns of progression in smoldering multiple nrc1098 1002/cncr.23783
myeloma. Nat Commun. 2018;9(1):3363. doi:10. 56. Manier S, Sacco A, Leleu X, Ghobrial IM, 65. Rajkumar SV, Gertz MA, Lacy MQ, et al.
1038/s41467-018-05058-y Roccaro AM. Bone marrow microenvironment in Thalidomide as initial therapy for early-stage
45. Mailankody S, Kazandjian D, Korde N, et al. multiple myeloma progression. J Biomed Biotechnol. myeloma. Leukemia. 2003;17(4):775-779. doi:10.
Baseline mutational patterns and sustained MRD 2012;2012:157496. doi:10.1155/2012/157496 1038/sj.leu.2402866
negativity in patients with high-risk smoldering 57. Bianchi G, Munshi NC. Pathogenesis beyond 66. Riccardi A, Mora O, Tinelli C, et al; Cooperative
myeloma. Blood Adv. 2017;1(22):1911-1918. doi:10. the cancer clone(s) in multiple myeloma. Blood. Group of Study and Treatment of Multiple
1182/bloodadvances.2017005934 2015;125(20):3049-3058. doi:10.1182/blood-2014- Myeloma. Long-term survival of stage I multiple
46. Walker BA, Wardell CP, Melchor L, et al. 11-568881 myeloma given chemotherapy just after diagnosis
Intraclonal heterogeneity is a critical early event in or at progression of the disease: a multicentre
58. Das R, Strowig T, Verma R, et al. randomized study. Br J Cancer. 2000;82(7):1254-
the development of myeloma and precedes the Microenvironment-dependent growth of
development of clinical symptoms. Leukemia. 2014; 1260. doi:10.1054/bjoc.1999.1087
preneoplastic and malignant plasma cells in
28(2):384-390. doi:10.1038/leu.2013.199 humanized mice. Nat Med. 2016;22(11):1351-1357. 67. Weber D, Rankin K, Gavino M, Delasalle K,
47. Papaemmanuil E, Gerstung M, Bullinger L, et al. doi:10.1038/nm.4202 Alexanian R. Thalidomide alone or with
Genomic classification and prognosis in acute dexamethasone for previously untreated multiple
59. Rasche L, Chavan SS, Stephens OW, et al. myeloma. J Clin Oncol. 2003;21(1):16-19. doi:10.
myeloid leukemia. N Engl J Med. 2016;374(23): Spatial genomic heterogeneity in multiple myeloma
2209-2221. doi:10.1056/NEJMoa1516192 1200/JCO.2003.03.139
revealed by multi-region sequencing. Nat Commun.
48. Bolli N, Biancon G, Moarii M, et al. Analysis of 2017;8(1):268. doi:10.1038/s41467-017-00296-y 68. Mateos MV, Hernández MT, Giraldo P, et al.
the genomic landscape of multiple myeloma Lenalidomide plus dexamethasone versus
60. Landgren O, Hofmann JN, McShane CM, et al. observation in patients with high-risk smouldering
highlights novel prognostic markers and disease Association of immune marker changes with
subgroups. Leukemia. 2017. doi:10.1038/leu.2017.344 multiple myeloma (QuiRedex): long-term follow-up
progression of monoclonal gammopathy of of a randomised, controlled, phase 3 trial. Lancet
49. Walker BA, Mavrommatis K, Wardell CP, et al. undetermined significance to multiple myeloma. Oncol. 2016;17(8):1127-1136. doi:10.1016/S1470-2045
A high-risk, Double-Hit, group of newly diagnosed JAMA Oncol. 2019. doi:10.1001/jamaoncol.2019.1568 (16)30124-3
myeloma identified by genomic analysis. Leukemia. 61. D’Arena G, Gobbi PG, Broglia C, et al; Gimema
2019;33(1):159-170. doi:10.1038/s41375-018-0196-8 69. Mateos MV, Hernández MT, Giraldo P, et al.
(Gruppo Italiano Malattie Ematologiche Lenalidomide plus dexamethasone for high-risk
50. Maura F, Petljak M, Lionetti M, et al. Biological Dell’Adulto); Multiple Myeloma Working Party; Gisl smoldering multiple myeloma. N Engl J Med. 2013;
and prognostic impact of APOBEC-induced (Gruppo Italiano Studio Linfomi) Cooperative 369(5):438-447. doi:10.1056/NEJMoa1300439
mutations in the spectrum of plasma cell dyscrasias Group. Pamidronate versus observation in
and multiple myeloma cell lines. Leukemia. 2018; asymptomatic myeloma: final results with 70. Korde N, Roschewski M, Zingone A, et al.
32(4):1044-1048. doi:10.1038/leu.2017.345 long-term follow-up of a randomized study. Leuk Treatment with carfilzomib-lenalidomide-
Lymphoma. 2011;52(5):771-775. doi:10.3109/ dexamethasone with lenalidomide extension in
51. Zhao S, Choi M, Heuck C, et al. Serial exome patients with smoldering or newly diagnosed
analysis of disease progression in premalignant 10428194.2011.553000
multiple myeloma. JAMA Oncol. 2015;1(6):746-754.
gammopathies. Leukemia. 2014;28(7):1548-1552. 62. Hjorth M, Hellquist L, Holmberg E, Magnusson doi:10.1001/jamaoncol.2015.2010
doi:10.1038/leu.2014.59 B, Rödjer S, Westin J; Myeloma Group of Western
Sweden. Initial versus deferred melphalan-

E8 JAMA Oncology Published online December 12, 2019 (Reprinted) jamaoncology.com

© 2019 American Medical Association. All rights reserved.

Downloaded From: https://jamanetwork.com/ by a Western University User on 12/12/2019

You might also like