Download as pdf or txt
Download as pdf or txt
You are on page 1of 20

Leukemia

https://doi.org/10.1038/s41375-019-0660-0

REVIEW ARTICLE

Multiple myeloma gammopathies

Extramedullary multiple myeloma


Manisha Bhutani1 David M. Foureau2 Shebli Atrash
● ●
1 ●
Peter M. Voorhees1 Saad Z. Usmani1

Received: 9 April 2019 / Revised: 31 July 2019 / Accepted: 12 August 2019


© The Author(s), under exclusive licence to Springer Nature Limited 2019

Abstract
Extramedullary multiple myeloma (EMM) is an aggressive subentity of multiple myeloma, characterized by the ability of a
subclone to thrive and grow independent of the bone marrow microenvironment, resulting in a high-risk state associated with
increased proliferation, evasion of apoptosis and treatment resistance. Despite improvement in survival for most patients
with multiple myeloma over recent decades, outcomes are generally poor when EMM develops. Understanding the
molecular underpinnings leading to homing of plasma cells in ecosystems outside the bone marrow will be crucial for
therapeutically manipulating the microenvironment and targeting key signaling pathways. Herein, we discuss the
evolutionary biology of EMM, underscore the importance of a uniform definition, discuss prognostic significance, and
1234567890();,:
1234567890();,:

provide current and emerging treatment strategies for managing this rare subentity of multiple myeloma.

Introduction myeloma, the diagnosis of EMM is typically made by the


presence of pathologic soft tissue masses on imaging
Multiple myeloma is characterized by clonal proliferation of [computed tomography (CT)-scan, PET/CT, magnetic
neoplastic plasma cells within the bone marrow resulting in resonance imaging (MRI), or ultrasound], biopsy or physi-
anemia, myelosuppression and bone destruction, as well as cal examination [7]. EMM is inherently a high-risk stage
clinical consequences from the paraproteinemia on kidney with a poor prognosis. The molecular mechanisms under-
function and other organ systems [1]. Extramedullary lying the development of EMM have not been fully defined.
multiple myeloma (EMM) is a less frequent manifestation Various cytogenetic abnormalities are observed, and few
of multiple myeloma, where myeloma cells become inde- studies have generated gene sequencing profiles in small
pendent of bone marrow microenvironment, infiltrate other patient cohorts that discriminate EMM from classic multiple
organs and/or circulate freely in the blood [2, 3]. Patients myeloma [8–11]. Treated the same way, the outcomes for
present with involvement of lymph nodes, skin, soft tissues, patients with EMM are different from multiple myeloma
central nervous system (CNS), thoracoabdominal organs, without extramedullary manifestations [11]. While PCL and
effusions, or any other anatomic sites, either at initial CNS EMM portend a poor prognosis, the outcomes can be
diagnosis (primary EMM) or at the time of relapse (sec- very heterogeneous for other manifestations. Sensitive
ondary EMM) [2, 4]. Rare cases evolve to or present with a imaging including PET/CT and MRI are important part of
leukemic phase termed plasma cell leukemia (PCL) [5], diagnosis and response evaluation. The purpose of therapy
which we consider as the most aggressive type of EMM as is to eradicate the myeloma clone from bone marrow and
discussed in this review. Occasionally there is involvement extramedullary compartments. We consider these points
of multiple extramedullary sites without bone marrow while reviewing the pathobiology, diagnosis, prognosis and
involvement [6]. In patients with confirmed multiple therapeutic options of EMM.

Controversies surrounding the definition


* Saad Z. Usmani
saad.usmani@atriumhealth.org
There is no uniform definition of EMM. A three-stage
1
Department of Hematologic Oncology and Blood Disorders, anatomic classification was first proposed in 1969 by Pas-
Levine Cancer Institute/Atrium Health, Charlotte, NC, USA mantier and Azar [12] based on review of 57 autopsied
2
Immune Monitoring Core Laboratory, Levine Cancer Institute/ cases of multiple myeloma: Stage I or intraskeletal, in
Atrium Health, Charlotte, NC, USA which myeloma nodules were confined to the skeleton;
M. Bhutani et al.

Stage II or paraskeletal, implying spread to adjoining tis- cells along the entire bone marrow compartment. The dis-
sues; stage III or extraskeletal, with gross spread to distant semination of myeloma cells from bone marrow ecosystem
sites. Earlier studies described a higher incidence of EMM to blood and distant tissues (organ-specific ecosystems)
in younger patients and in those with IgD myeloma and involves multiple processes. The precise sequence of events
nonsecretory myeloma [13, 14]. The definitions of EMM and the extent to which these processes are activated at
vary greatly. Some groups restrict the definition solely to various stages of extramedullary progression remain poorly
soft tissue masses in extraosseous locations resulting from understood; however, few principles have emerged that
hematogenous spread, referred to here as extramedullary- unify our understanding of biologic evolution to a high- risk
extraosseous (EM-E) [4, 7, 11]. Others have used a broader state of EMM and PCL.
definition inclusive of bone-related plasmacytomas that The primary genetic events endow tumor-initiating
extend via disruption of cortical bones into contiguous soft abilities in a clone of plasma cells critical for progression
tissues, referred to in this paper as extramedullary-bone from precursor states to active multiple myeloma, while
related (EM-B) [15]. It is important to differentiate between acquisition of secondary and terminal genetic events, in
the two types, as the outcome is worst for patients with EM- collaboration with microenvironment influence, intra and
E compared with those with EM-B [16, 17]. Moreover, extracellular signaling, epigenetic changes and immune
studies suggest that plasma cells extending from the bone evasion, potentially trigger egress to ecosystems outside the
into the adjacent soft tissues are simply an extension of the bone marrow (Fig. 1). Plasma cells that invade into the
same “biological compartment” as the bone itself, whereas marrow neo-vasculature or into the vasculature of adjacent
plasma cells at distant anatomical sites carry different bio- normal tissues encounter physical challenges en route to
logical characteristics [4, 18]. Importantly, the definition of distant tissues, which include failure to attach to a substrate,
EMM must explicitly exclude ‘solitary extramedullary hydrodynamic flow, and shear stress. In addition, immune
plasmacytoma’ and ‘solitary bone plasmacytoma’ (Table 1) interactions can target circulating malignant cells for elim-
[19, 20]. PCL can be considered an extreme variant of ination. Once in the circulation, few plasma cells manage to
aggressive EMM characterized by rapid progression, drug extravasate, seed into new tissue microenvironment, adapt
resistance and short survival [5]. Even lower levels of cir- to survival signals and colonize into the extramedullary
culating clonal plasma cells, with a cutoff ≥5% portend a ecosystem.
similar adverse prognostic impact as traditionally defined In the following sections, we focus on the molecular
PCL [21]. Although PCL fulfills the definition of EMM, mechanisms and adhesion molecules that play a role in the
some authors reason that PCL is a well-defined pathologic pathogenesis of EMM. Epigenetic and immune changes
entity and should be excluded from the EMM spectrum [7]. also play a role, but these factors have been studied less
We recommend restricting the definition of EMM to the extensively.
presence of extramedullary disease in a patient meeting the
definition of multiple myeloma, excluding those with soli-
tary extramedullary/bone plasmacytoma (Table 1). To have Cytogenetic and molecular hallmarks
a better understanding of the behavior of contiguous versus
hematogenous spread, it is important to collect prospective The cytogenetics, FISH, gene expression profiling (GEP)
data in EMM with respect to EM-E and EM-B subtypes. and deep sequencing studies comparing samples taken from
We propose that both secondary and primary PCL be different individuals who have multiple myeloma, EMM or
included in the definition of EMM as it is not uncommon PCL have identified gradual accumulation of genetic events
for these patients to have concomitant or subsequent as the disease transitions to a more aggressive phenotype. In
extraosseous involvement of liver, spleen, lymph nodes, the context of a disease model where EMM and PCL are
and other soft tissues [5, 22, 23]. considered as one extreme of the myeloma spectrum, mul-
tistep transformation includes acquisition of sequential
genetic hits leading to generation of branching evolutionary
Evolutionary biology pathways, with better adaptation of clonal cells to their
microenvironment leading to ‘fitter’ clones that outcompete
There are important biological differences between multiple dominant clones [24]. Spatiotemporal comparisons of bone
myeloma and EMM that might explain the propensity for marrow and targeted biopsies of EMM and multiple focal
dissemination and a more aggressive clinical course in the lesions reveal enrichment of pathways in EMM sites that
latter. Multiple myeloma cells primarily grow in the bone were not identified in original bone marrow biopsies, sup-
marrow and are dependent upon the microenvironment for porting a model for progression with clonal sweeps in the
their growth and survival. The presence of multiple bone early phase and regional evolution in advanced disease [25].
lytic lesions suggests an efficient trafficking of myeloma The time at which two subclones evolve at extramedullary
Table 1 Defining features of EMM and other relevant plasma cell dyscrasias
Plasma cell dyscrasia Definition Comments

Extramedullary multiple • Presence of extramedullary disease in a patient meeting the definition of Diagnosis can be made by imaging (PET/CT and/or MRI) in a patient with
myeloma (EMM) multiple myeloma confirmed multiple myeloma either at the time of diagnosis (primary EMM)
• Diagnostic (IMWG) criteria for multiple myeloma must be fulfilled or at relapse/progression (secondary EMM).
• Does not include solitary extramedullary plasmacytoma or solitary bone Pathologic confirmation is recommended wherever possible.
plasmacytoma (discussed below)
Extramedullary multiple myeloma- • EMM involving soft tissue or viscera in extraosseous locations resulting Examples include involvement of lymph nodes, liver, pleura, testis, skin,
extraosseous (EM-E) from hematogenous spread, not contiguous to involved bone CNS, kidneys, blood.
Extramedullary multiple myeloma

Extramedullary multiple myeloma- bone • Extramedullary plasma cell mass arising from the underlying bone (e.g. Some authors do not consider EM-B in the definition of EMM.
related (EM-B) ribs, vertebrae, skull, sternum, pelvis) with extension to contiguous To have a better understanding of the behavior of contiguous versus
paraskeletal area or soft tissue via cortical disruption hematogenous spread, we believe it is important to collect prospective data
in patients with EMM with respect to both EM-E and EM-B subtypes.
Solitary extramedullary plasmacytoma • A distinct diagnostic entity that must not be confused with EMM. Most common site is upper aerodigestive tract, but other sites have been
• Biopsy -proven localized monoclonal plasma cell mass localized to a described such as gastrointestinal tract, urinary bladder, thyroid, breast,
SINGLE extramedullary site in the absence of other defining events for testes, parotid glands or lymph nodes.
multiple myeloma Treatment involves surgical resection and/or radiotherapy. Estimated
• No evidence of clonal plasma cells in bone marrow disease-free survival, about 80% at 10 years.
• No more than one primary lesion on low dose whole-body CT, PET/CT
or MRI of the whole body or of spine and pelvis
• Absence of end-organ damage (CRAB) or systemic amyloidosis that can
be attributed to the plasma cell proliferative disorder
Solitary bone plasmacytoma • A distinct diagnostic entity that must not be confused with EMM The common affected sites are vertebra, pelvis, ribs, upper extremities, face,
• Biopsy -proven localized monoclonal plasma cell mass localized to a skull, femur, or sternum.
SINGLE bone with or without extension to adjacent soft tissue, in the The primary treatment is definitive local radiation therapy. Surgical
absence of other defining events for multiple myeloma. intervention may be necessary in patients with vertebral instability or
• No evidence of clonal plasma cells in bone marrow (or <10% in the case pathologic fracture of a long bone.
of solitary bone plasmacytoma with minimal marrow involvement) Estimated disease-free survival about 50% at 10 years.
• No more than one primary lesion on low dose whole-body CT, PET/CT Patients with minimal marrow involvement (up to 10% plasma cells) treated
or MRI of the whole body or of spine and pelvis like solitary bone plasmacytoma have a higher risk of progression to
• Absence of end-organ damage (CRAB) or systemic amyloidosis that can symptomatic myeloma with a 60% chance of recurrence or progression
be attributed to the plasma cell proliferative disorder. within three years.
Patients with SPB with 10% or more clonal plasma cells in the bone
marrow are considered to have multiple myeloma and treated as such.
Primary plasma cell leukemia • An extreme variant of EMM, defined by the presence of 20% and/or an We propose both secondary and primary PCL be included in the definition
absolute number >2 × 10e9/L of (clonal) plasma cells in the peripheral of EMM.
blood without a prior history of multiple myeloma
• Even lower levels of circulating clonal plasma cells, with a cutoff ≥5%
portend a similar adverse prognostic impact as traditionally defined PCL,
indicating that a lower-cut off may be adapted in future consensus
• Clonality establishment by flow-cytometry must be given consideration,
as polyclonal plasma cells can be detected in blood in other conditions
Secondary plasma cell leukemia • Plasma cell leukemia, which occurs in a patient with a previously
diagnosed multiple myeloma, generally as a progressive/transformation
event during relapsed/refractory course of illness.
M. Bhutani et al.

Fig. 1 Model illustrating central drivers in pathogenesis of EMM and marrow ecosystem with complex network signals emanating from
plasma cell leukemia. Genetic and microenvironment mechanisms are plasma cells and various components of their microenvironment, such
important for biologic evolution to a high-risk state of EMM and PCL. as osteoclasts, endothelial cells, stromal cells, and cells of the immune
Primary immunoglobulin heavy chain (IgH) translocations with five system. The balance is delicate and tightly regulated. A variety of
recurrent chromosomal partners (4p16, 6p21, 11q13, 16q23, and genetic (and epigenetic) changes endow critical phenotype traits to
20q11) and hyperdiploidy (typically with trisomies of chromosomes 3, malignant plasma cells that allow territorial expansion to outside
5, 7, 9, 11, 15, 19, and 21) are considered initiating genetic events that organ-specific ecosystems, via proliferative self-renewal, adhesion,
endow tumor-initiating abilities in a clone of plasma cells critical for angiogenesis, migration, and invasion. Important chemokines, adhe-
progression from precursor states to active multiple myeloma. sion molecules and growth factors crucial to each step are shown in the
Acquisition of secondary chromosomal abnormalities (such as dele- green box. The process of clonal evolution is not as simple, and the
tions of chromosomes) or terminal events (secondary chromosomal exact sequence of events is not known. Progression is also accom-
translocations and mutations involving individual genes) are critical to panied by altered interactions of the tumor cells with various com-
progression to aggressive forms of multiple myeloma, such EMM or ponents of their microenvironment, such as osteoclasts, endothelial
plasma cell leukemia. The evolution takes place within the bone cells, and cells of the immune system

sites may be temporarily synchronous (primary EMM) or of a clone that can both proliferate and self-renew. Some
develop at different time intervals (secondary EMM). translocations confer high-risk features, for example, t(4;14)
The primary genetic events such as hyperdiploidy, del leads to epigenetic dysregulation via expression of MMSET
(13)q(14) and recurrent translocations of immunoglobulin [28], and t(14;16) and t(14;20) cause an aberrant tran-
heavy chain (IgH) loci with five chromosomal partners scriptional program via overexpression of the oncogenes
(4p16, 6p21, 11q13, 16q23, and 20q11) are typically clonal MAF and MAFB, respectively. Secondary acquired events
[24, 26], and lead to upregulation of cyclin D1, D2, and D3 (amplification of 1q21 ≥ 4 copies and loss of 17p) or late
thereby triggering G1/S checkpoint cell cycle progression terminal events (MYC translocation or 8q24 copy number
and downstream driving of cellular proliferation [27]. changes, mutation of the MAPK signaling pathway or the
Consequently, there is phenotypic transition and evolution nuclear factor-κB pathway) occur in a divergent manner at
Extramedullary multiple myeloma

subclonal level during disease progression and cause in 14 patients with EMM using DNA sequencing for a
enhanced DNA damage and genome instability. The high- targeted panel of 50 tumor suppressor and oncogenes that
risk features alter tumor cell biology and allow propagating revealed a high frequency of activating RAS mutations in
cells to cross the bone marrow boundary leading to 67% bone marrow samples (6/9 patients) and in 64%
aggressive forms of multiple myeloma, such as EMM and extramedullary samples (7/11 patients). The incidence of
PCL (Fig. 1) [26]. Compared with classic multiple mye- RAS mutations in this cohort was close to PCL [43] but
loma, EMM and PCL are molecularly distinguished by a much higher than previously reported 23–44% in newly
high prevalence of high-risk chromosomal abnormalities, a diagnosed and relapsed multiple myeloma [44]. In three of
more complex genomic profile, and immunophenotypic six patients with identical IgH sequences in intramedullary
features related to adhesion and chemokine molecules and extramedullary plasma cells, RAS mutations were only
expression (Table 2) [4, 9, 10, 29–37]. observed in plasma cells from extramedullary sites, thus
suggesting a role of RAS mutations in transition to EMM
EMM [37]. Egan et al. reported an acquired truncating mutation of
cereblon as well as point mutations in proteasome subunit
Small case series have described cytogenetic/FISH features G2 and the glucocorticoid receptor as an explanation of
in diagnostic marrow samples of patients who subsequently drug resistance in a patient presenting with progressive,
relapse/progress with EMM, In a series of 19 patients who multi-drug refractory EMM [45]. Whole-exome, whole-
progressed to EMM, the most frequent cytogenetic genome and RNA sequencing of a biopsy taken from the
abnormality at diagnosis was 13q deletion, seen in 11/19 neck mass revealed a highly disturbed genome including
cases followed by t(4;14) in 10/19 (52%) and deletion 17p 271 nonsynonymous, somatic point mutations in genes
in 4/19 (21%) [8]. In a retrospective study of 117 multiple including KRAS, PIK3CA, ATM, and NFKB2, suggesting
myeloma patients treated on bortezomib-based protocols, the role of these candidate genes in extramedullary trans-
the presence of t(11;14), del 13/13q by FISH and del(17p) formation and potential exploitation with MEK and PI3K
at diagnosis did not predict for development of EMM at inhibitors.
relapse, but all patients who did develop EMM lacked t
(11;14) at diagnosis [38]. Evaluation of cytogenetic differ- PCL
ences between multiple myeloma patients with and without
EM-E at presentation showed particularly high incidence of Although a higher frequency of t(11;14) using FISH probes
p53 deletion in EM-E, 34.5% (10/29) vs. 11.9% (29/243), for targeted sequences is identified in primary PCL
respectively [32]. A case report described a newly acquired (Table 1), this abnormality often associated with other high-
TP53 deletion in the EM-E lesion, but wild type TP53 in the risk molecular events explain an inferior outcome for PCL
bone marrow with a marked dissociation between response in contrast to favorable prognosis associated with this iso-
obtained in bone marrow and that achieved in soft tissue lated abnormality seen in multiple myeloma [9, 22].
[39]. Although the reported incidence of TP53 mutation is Number and patterns of somatic variants in PCL are
10-30% in marrow localized multiple myeloma, it was higher and more complex than multiple myeloma. Com-
detected in tissues from 8/9 (89%) patients with CNS EMM parison of GEP of patients with primary PCL (n = 20) and
[40]. In another series of 12 patients, 75% of EMM biopsy multiple myeloma (n = 1096) treated on ‘total therapy’ (TT)
specimens revealed increased p53 immunostaining, whereas protocols revealed a signature of 203 genes predominantly
only 8% of bone marrow specimens showed nuclear accu- belonging to the lipid-metabolism and myeloid differentia-
mulation of p53 [41]. Thus the tumor suppressor gene TP53 tion pathway that distinguished primary PCL and non-PCL
seems to be an important driver associated with EMM. cases [36]. PET data, available at baseline for 724 patients,
Analysis of extramedullary tissue biopsies in 19 patients revealed a higher incidence of extramedullary involvement
with EM-E and 11 with EM-B revealed no significant dif- in the primary PCL group than in multiple myeloma (21%
ference in recurrent cytogenetic abnormalities between the versus 6%; P = 0.05). In another GEP study, primary
two types: del(17p13) 32% vs. 27%, del(13q14) 35% vs. immunoglobulin translocations were identified in 63% (12/
27%, MYC-overexpression 28% vs. 18% and t(4;14) 37% 19) of patients with PCL, including MAF t(14;16) in 32%,
vs. 18%, respectively [42]. t(14;20) in 10%, t(11;14) in 16%, and t(4;14) in 10% [46].
Using GEP by both 70-gene and 80-gene models, EMM Furthermore, 32% (6/19) of patients had at least one MYC
was linked to high-risk molecular subgroups defined by the translocation, which are known to strongly correlate with
MF subgroup often associated with t(14;16) and t(14;20), disease progression. In another cohort of patients, whole-
and the PR subgroup representing highly proliferative dis- exome sequencing on primary PCL cells identified 14
ease [11]. Haart et al. [37] characterized molecular altera- candidate cancer driver genes involved in cell-matrix
tions between paired bone marrow and soft tissue biopsies adhesion and membrane organization (SPTB, CELA1), cell
Table 2 Important biologic features that distinguish EMM and primary PCL from multiple myeloma
Characteristics Multiple myeloma EMM Primary PCL References

Immunophenotype of aberrant Bone marrow plasma cells: Positive or Extramedullary site plasma cells: Positive or strong Circulating plasma cells: Positive or strong Jelinek et al. [29], Jelinek et al.
plasma cells strong positive expression of CD28, CD33, positive expression of CD44, CD81, nestin positive expression of CD19, CD20, CD23, [30], Rihova et al. [31]
CD38, CD56, CD117, CD138, BCMA, CS1 Negative or weak expression of CD19, CD27, CD44, CD45.
(CD319), and CD200. CD56, CD117 Negative or weak expression of CD27, CD56,
Negative or weak expression of CD19, CD71, CD117, and HLA-DR.
CD20 CD27, CD45, and CD81 Expression of CD23 has been associated with t
(11;14)
Adhesion molecule, chemokine Adhesion molecule downregulation (CD56, P- Surface integrin downregulation (CD11a, Reviewed in Weinstock and
receptors selectin, VLA-4) CD11c, CD29, CD49d, CD49e) Ghobrial [4] and Jelinek et al.
Migration molecule upregulation (CD44 isoforms) Adhesion molecule downregulation (CD33, [29]
Angiogenesis upregulation (VEGF, MMP-9, CD56, CD117, CD138)
angiopoietin-1, CD31) Activation molecules downregulation (CD28,
Chemokine receptor downregulation (CCR1, CCR2, CD38, CD81)
CXCR4/SDF-1alpha)
Cytogenetic abnormalities Chang et al. [9], Deng et al. [32],
Royer et al. [33], Avet-Loiseau
et al. [34]
t(11;14) 13% (19/145) 14% (4/28) 27% (6/22)
t(4;14) 13% (24/179) 23% (7/31) 29% (8/28)
t(14;16) 3% (32/1003) 8% (2/26) 17% (11/70)
del(13q) 41% (65/158) 59% (17/29) 63% (22/35)
del(17p) 11% (17/155) 34.5% (10/29) 37% (13/35)
Amp (1q21) 34% (45/133) 54% (14/26) 51% (20/39)
Del (1p21) 18% (36/203) 33% (13/40)
GEP and NGS mutational panel Neri et al. [35], Usmani et al.
[11], Usmani et al. [36], Cifola
et al. [10], de Haart et al. [37]
TP53 3.1% (4/129) 25% (6/24)
DIS3 18.5% (24/130) 25% (6/24)
FAM46C 11.7% (15/128) 4.2% (1/24)
NRAS 26.5% (35/132) 4.2% (1/24)
KRAS 32.6% (43/132) 16.7% (4/24)
BRAF 10.6% (14/132) 20.8% (5/24)
GEP 70 high risk 19% (216/1140) 46% (12/26) 44% (7/16)
GEP 80 high risk 13.7% (156/1140) 42% (11/26) 31% (5/16)
Mutations in TP53, RB1, KRAS, FAK have been Lipid-metabolism and myeloid differentiation
described in up to 50% of patients with EMM genes overexpressed: (CD14(cell membrane
Chr 17p deletion and nonhyperdiploid status are both LPS receptor), TRAF2 and CCL2).
over-represented in EMM Enrichment of Cadherin/Wnt signaling, ECM-
Angiogeneis related genes overexpressed receptor, and G2/M cell cycle checkpoint
(angiopoietin 1, SPARC, NOTCH3, thrombospondin
2, TIMP3, PDGF-alpha, PDGF-beta, and
fibronectin1)
Adhesion molecule genes overexpressed (CD31,
ENG, SPARC)
M. Bhutani et al.
Extramedullary multiple myeloma

cycle and apoptosis (CIDEC), genome stability (KIF2B), dehydrogenase A has been associated with egress of plasma
RNA metabolism (DIS3, RPL17), and protein folding cells to extramedullary sites [58]. Focal adhesion kinase
(CMYA5) being the most affected, with enrichment of (FAK), a protein whose activation can affect cell survival,
functional pathways, including cadherin and Wnt signaling, migration, and invasion via interaction with the phosphatase
extracellular matrix (ECM)-receptor interaction, ECM and tensin homolog (PTEN) pathway has shown to play a
organization, and G2/M cell cycle checkpoint [10]. role in EMM. In a series of 55 MM patients, 12 with EMM
had significantly higher expression level of FAK mRNA in
Role of adhesion and chemokine molecules their bone marrow nuclear cells in contrast to those without
EM infiltration [59].
The multiple myeloma cells within the bone marrow eco- Angiogenesis is a central component of the bone marrow
system engage with stromal cells, endothelial cells, hema- vascular microenvironment. Experiments have shown that
topoietic cells, immune cells, osteoblasts, osteoclasts, cells within the marrow release soluble “angiocrine” factors
extracellular matrix, and liquid milieu to facilitate autocrine that promote plasma cell expansion. Bone marrow stromal
and paracrine signaling [47, 48]. Adhesion of myeloma cells from multiple myeloma patients express multiple
cells to bone marrow stromal cells is mediated by mole- proangiogenic molecules such as vascular endothelial
cules, including CD44 isoforms, neuronal adhesion mole- growth factor (VEGF), angiopoietin 1 (Ang-1), basic-FGF
cule (NCAM), beta-1 integrins [very late antigen (VLA) 4, (bFGF), platelet-derived growth factor (PDGF), hepatocyte
VLA5] and beta-2 integrins [leukocyte function-associated growth factor (HGF), transforming growth factor-alpha
antigen 1 (LFA-1), intercellular adhesion molecule (TGF-α), and IL-1 [60]. Many genes involved in angio-
(ICAM1)] and chemokine receptors (Fig. 1) [49]. Adhesion genesis, such as TIE2, NOTCH3, and several factors that
favors secretion of cytokines, chemokines and other factors promote angiogenesis, including VEGF, matrix
that promote angiogenesis and an immune suppressive metalloproteinase-9 (MMP-9), Ang-1, and CD31, appear to
milieu. be upregulated in EMM compared with MM [61].
A significant effort has been devoted to finding mole-
cules that direct plasma cells to extramedullary sites. Stu-
dies have shown that plasma cells derived from EMM are Incidence
characterized by decreased expression of CD56, the beta 5
integrin adhesion molecule [50–52] and an increased The reported incidence of EMM is generally underestimated
expression of CD44 isoforms involved in cell proliferation given the inconsistencies in definitions and method of eva-
and migration (Table 2) [51]. Specifically, expression of luation. Most studies predated the era of novel therapies and
CD44 variant isoform 6 (CD44v6) and CDv9 are associated more routine application of sensitive imaging modalities
with plasma cell motility and poor prognosis in multiple (Table 3) [8, 11, 15, 16, 32, 38, 62–69]. The reported inci-
myeloma [53, 54]. Decreased expression of other adhesion dence of EM-E at diagnosis ranges from 1.7 to 4.5%
molecules (P-selectin, VLA-4) has also been associated [11, 66, 67]. EM-B involvement is more frequent and varies
with EMM in various murine model studies [55]. It remains from 7 to 34.2% [2, 15]. At relapse, the incidence of EM-E
unclear whether loss of these receptors alters the adhesive ranges from 3.4 to 10% while EM-B involvement from 6 to
potential of the tumor cell and/or allows escape from 34% [15, 16, 38]. In the largest series evaluating 3744
immune surveillance. The chemokine receptor CXCR4 is a multiple myeloma patients from a European registry, 14.5%
transmembrane, G-protein coupled receptor that is expres- had EM-B and 3.7% had EM-E at diagnosis [63]. Hypo-
sed on both normal and malignant plasma cells and is thetical concerns have been raised about an increased inci-
involved in plasma cell homing to the bone marrow where dence of secondary EMM in the era of novel agents, however
its ligand SDF-1 (also known as CXCL12) is secreted by whether this is truly a consequence of exposure to novel
osteoblasts and stromal cells. Impairment of SDF-1/ agents is contentious, as some studies have shown an
CXCR4 signaling has been associated with extramedullary increased risk with these agents [70, 71], whereas others have
transformation [2]. Concomitant downregulation of CXCR5 not [15, 38]. Currently, there are no evidence-based data to
and CCR7 have shown to promote plasma cell motility by support that the risk of EMM is influenced by the choice of
decreasing their sensitivity to B and T zone chemokines the initial treatment. As patients are being followed‐up over
CXCL13, CCL19, and CCL21 [56]. In addition, an upre- longer periods of time, their probability of eventually relap-
gulation of epithelial–mesenchymal transition-like tran- sing with EMM increases. Autopsy studies in the past
scriptional activity that leads to higher CXCR4 expression revealed the presence of extraskeletal involvement in
may play role in extramedullary dissemination [57]. Bone approximately 70% of patients, suggesting that EMM
marrow hypoxia, which induces a metabolic switch to represents a natural evolution of the disease [12]. A higher
aerobic glycolysis via upregulation of HIF1alpha and lactate incidence of EMM has been reported in patients after
Table 3 Incidence and survival of EMM in select retrospective studies
Incidence of EMM No. of MM Diagnostic strategy Treatment PFS (EMM OS (EMM vs. MM) Remarks Reference
patients (period vs. MM)
covered)

11% primary EMM 2332 Imaging per study Three trials for transplant- Median PFS (25.3 Median OS (63.5 vs. This is a meta-analysis of Montefusco
(91% EM-B and protocol; skeletal survey, eligible, and five trials for vs. 25.2 mo). 79.9 mo). 5-y OS eight treatment trials. et al. [62]
4% EM-E) MRI, or CT. And/or transplant-ineligible 5-y PFS (19% vs. (51% vs. 59%, p = In multivariate analysis
physical examination. patients. 22%, p = 0.46) 0.01) the presence of EMM
Three trials included an did not impact PFS,
IMiD (lenalidomide in while it was associated
almost all cases), three trials with a reduced OS (HR
a PI, and four trials both. Six 1.41, 95% CI 1.16-1.71;
out of eight trials-included p < 0.001).
maintenance.
14.5% primary EM-B, 3744 Patients who had full data All patients received an up- EM-B: 3-y PFS EM-B: 3-y OS (77.7% Between 2005 and 2014, Gagelmann
3.7% primary EM-E (2005–2014) available on front single ASCT within (50% vs.47.9%, p vs. 80.1%, P = 0.09) the EMM incidence per et al. [63]
extramedullary 12 months of diagnosis or a = 0.78) EM-E: 3-y OS (58% year increased from
involvement (yes or no) tandem ASCT within EM-E: 3-y PFS vs. 80.1%, P < 0.001) 6.5% to 23.7%.
at time of diagnosis, its 6 months from first ASCT (39.9% vs. 47.9%,
location, and the number as first-line therapy. P = 0.001)
of sites were included.
14% 456 Skeletal survey, whole Induction included first- Median OS 3.1 y Secondary plasma cell Mangiacavalli
primary EMM (2000–2010) body low dose CT, generation novel agents primary EMM, leukemia was also et al. [64]
28% secondary EMM MRI, PET/CT (Thalidomide, 2 y secondary EMM included in the definition
(36% EM-B, 52% EM-E, Lenalidomide, Bortezomib), vs. 11 y p < 0.001) of secondary EM-E
both 12%) and ASCT EM-E vs. EM-B (1.6 y
vs. 2.4 y, P = 0.006)
16.2% primary EMM 271 MRI, PET/CT, and/or Induction using novel Median PFS (18 vs. Median OS (32 vs. Kumar et al.
biopsy/cytology agents, VAD, or alkylating 44 mo, P < 0.001) 100 mo, P < 0.001) [65]
agents. All patients
underwent ASCT.
4.8% primary EMM 834 Xray, ultrasound, CT, Induction therapy with Median TTP (11.5 Median OS (16.5 vs. Deng et al. [32]
3.4% secondary EMM (1993–2013) physical exam. Histologic novel agents-based vs. 25 mo, 40 mo, P < 0.001)
confirmation was regimens (Thalidomide, P < 0.001)
performed whenever Bortezomib or
possible. Lenalidomide), alkylating-
based regimens, or
dexamethasone-based
regimens. After induction
only five patients (12.5%)
received ASCT.
32.5% primary EMM-B, 117 Radiological imaging Eight clinical trials of first- Secondary EMM: PCL was also classified Varga et al. [38]
1.7% primary EM-E (2003–2012) (CT, PET/CT, MRI), line treatment with Median OS was 0.9 y as EMM.
27.4% secondary EM-B, biopsy, and/or physical bortezomib-based regimens, (0.1–4.8 y) for EM-E. The rate of development
16.2% secondary EM-E examination. with or without of secondary EMM at
M. Bhutani et al.
Table 3 (continued)
Incidence of EMM No. of MM Diagnostic strategy Treatment PFS (EMM OS (EMM vs. MM) Remarks Reference
patients (period vs. MM)
covered)

lenalidomide. Throughout Median OS was 2.47 y truncated follow up of


the follow-up period, 57 (0.1–8.7 y) for EM-B 5 y and 7 y was not
patients (48·7%) received an influenced by induction
ASCT and four patients therapy (bortezomib with
(3·4%) received an alloSCT lenalidomide vs. without
Extramedullary multiple myeloma

lenalidomide)
8.3% secondary EMM-E 663 Pathologic or radiologic All 55 patients with EMM Median OS from Weinstock et al.
(2005–2011) evidence of EMM at any were treated with a median EMM diagnosis was [66]
time following initial of four different regimens 1.3 y (0.8–2.3 y)
diagnosis. prior to development of
EM-B were excluded EMM. All underwent ASCT
from definition of EMM. and 15 also underwent
Only patients with alloSCT
biopsy-proven clonal
plasma cell infiltrates
were included.
14% secondary EMM-E. 226 Ultrasound, CT, or MRI. Conventional Median TTP after Median OS from Pour et al. [16]
10% secondary EMM-B (2005–2008) Biopsies were carried out chemotherapy, novel agents EM relapse was diagnosis (38 vs. 109
if the lesion was (Thalidomide or Bortezomib 5.4 mo. mo, P < 0.001)
accessible. containing regimens), Median OS EM-E vs.
ASCT, Interferon alfa. EM-B (30 vs. 45 mo,
P = 0.022)
Primary EMM: 2.41% of 1965 Baseline PET/CT ASCT within the context of Primary EMM: 5-y Primary EMM: 5-y OS The cumulative Usmani et al.
TT, 4.35% of non-TT, (2000–2010) performed at diagnosis TT protocols, non-TT PFS (21% vs. 50%, (31% vs. 59%, P < incidence of EMM was [11]
and 4.5% of non- for all patients protocols, and non-protocol P < 0.001) 0.001) significantly increased in
protocol patients. patients. patients who had GEP-
Secondary EMM: 3.43% defined high-risk disease
of TT, 5.2% of non-TT, at baseline and baseline
and 7.24% of non- cytogenetic
protocol patients abnormalities.
6.7% secondary EMM 357 Cytologically proven ASCT, ASCT-alloSCT, Median PFS of Median OS of CNS involvement in 5/ Rasche et al. [8]
(2007–2010) malignant effusion, or a alloSCT, novel agents. 2 months (95% CI 7 months (95% CI 24 cases
biopsy-proven plasma Treatments post EMM 0.08–3.92) 3.56–10.43)
cell tumor outside the included RT, dose-intense
bone marrow, and not chemotherapy including
originating from skeletal novel agents, and ASCT-
structures alloSCT.
9.2% secondary EMM-E. 174 PET/CT, MRI. Within the context of a Not provided Median OS This study specifically Short et al. [67]
(2007–2010) Of note, EM-B were phase II trial of (16 months versus not assessed the incidence of
excluded pomalidomide and low-dose reached, P = 0.002) EMM that develops
dexamethasone during the progression of
Table 3 (continued)
Incidence of EMM No. of MM Diagnostic strategy Treatment PFS (EMM OS (EMM vs. MM) Remarks Reference
patients (period vs. MM)
covered)

MM in patients who did


not have EMM at
diagnosis
7% primary EMM (85% 1003 MRI, PET, X-ray, Conventional chemotherapy Median PFS (18 vs. Median OS (36 vs. 43 Most patients (64 of 76, Varettoni et al.
EM-B and 15% EM-E) (1971–2007) physical examination (1971–1993), up front 30 mo, P = 0.003) mo, P = 0.36) 84%) had a single [15]
6% secondary EMM ASCT for age <65 year plasmacytoma.
(1994–1999), novel agents By time-dependent
(2000–2007) analysis, the presence of
EMM at any time
(primary or secondary)
was associated with
significantly shorter OS
(HR 3.26, P < 0.0001)
and PFS (HR 1.46, P =
0.04).
In the 208 MM patients
treated with ASCT, the
presence of EMM did
not significantly affect
PFS (P = 0.08) and OS
(P = 0.17).
20.4% secondary EMM 172 Sequential ASCT-alloSCT. Median PFS after 1-y OS after alloSCT Minnema et al.
Treatment of EMM included alloSCT not (61% vs. 73% for [68]
DLI, radiotherapy, or significantly relapsed MM without
chemotherapy different between EM) disease,
patients with and P = 0.494)
without EMM at
relapse
14.2% secondary EMM 70 Imaging guided by ASCT-alloSCT or alloSCT NA NA Of 70 patients included Perez-Simon
(1999–2004) symptoms with reduced-intensity in the study, 27 patients et al. [69]
conditioning relapsed (of which 10
were EMM).
EMM extramedullary multiple myeloma, EM-E extraosseous EMM, EM-B bone-related EMM, MM multiple myeloma, IMiD immunomodulatory drug, PI proteasome inhibitor, VAD (vincristine,
doxorubicin, dexamethasone), ASCT autologous stem cell transplant, alloSCT allogeneic stem cell transplant, TT total therapy, RT radiation therapy, PFS progression-free survival, OS overall
survival, TTP time to progression, y years, mo months, CI confidence interval, NA not available
M. Bhutani et al.
Extramedullary multiple myeloma

allogeneic stem cell transplantation (alloSCT) than auto- confirmed MM progression/relapse was associated with a
logous stem cell transplantation (ASCT) [68, 69, 72, 73]. median OS of 5 months for EM-E and 12 months for EM-B
Graft versus myeloma effect may not be as effective in cer- [16]. In a retrospective series, 226 patients with EMM
tain extramedullary sites due to lower infiltration of tumor- between 2010 and 2017 were analyzed [76]. Among the
specific lymphocytes into these sites relative to the bone primary EMM who underwent ASCT, the median PFS was
marrow. Another possibility is that patients with high-risk 38.9 months for EM-E and 51.7 months for EM-B (p =
features and multiply relapsed advanced disease are more 0.034), whereas OS was 46.5 months for EM-E and not
likely to undergo alloSCT. reached for EM-B (p = 0.002). However, for secondary
An important question is whether increased use of PET/ EMM, PFS was 11.4 months for EM-E and 20.9 months for
CT imaging would detect more cases of EMM. In few EM-B (p = 0.249), whereas OS was 13.6 months for EM-E
studies incorporating PET/CT at diagnosis, the reported and 39.8 months for EM-B (p = 0.093). The presence of
incidence of EMM from 3.4 to 10% seems not to reflect an EMM at certain anatomic sites such as blood, CNS, liver,
increase [11, 67, 74]. The recent International Myeloma lung, and muscles has been associated with earlier deaths
Working Group guidelines recommend the use of PET/CT than when these sites were not involved. EMM with mul-
for both newly diagnosed and relapsed/refractory multiple tiple involved sites carries a poor prognosis. Patients with
myeloma to determine the extent of bone damage and multiple site EMM had shorter 3-year PFS after first-line
extramedullary involvement [75]. PET/CT is being ASCT, 22.7% versus 49.4%, than with one involved site or
employed in several novel therapy studies. As results of multiple myeloma (p = 0.001), but both one and multiple
prospective studies become available, the role of PET/CT in involved sites showed worse 3-year OS in comparison to
determining the true incidence of EMM will be subject to patients with multiple myeloma [63]. The prognosis for
continual reassessment. patients who have PCL is extremely poor. An analysis of
the Surveillance, Epidemiology, and End Results database
that included 445 patients with primary PCL diagnosed
Prognosis between 1973 and 2009 reported median OS of 5, 6, 4, and
12 months, during the time periods of 1973–1995,
Tumor burden is generally high in patients with EMM, and 1996–2000, 2001–2005, and 2006–2009, respectively [77].
they usually present with severe anemia, thrombocytopenia,
hypercalcemia, and high rates of renal impairment. Elevated
serum lactate dehydrogenase, serum β2 microglobulin Current and emerging treatment strategies
level ≥ 5.5 g/dL and light chain escape are also typical fea-
tures of EMM, often associated with plasmablastic mor- Given the relative infrequency of EMM, exclusion of
phology or PCL [7]. Historically, the outlook for patients patients with nonsecretory EMM, PCL, and CNS myeloma
with EMM has been poor, with a median OS of <1 year for from clinical trials, as well as lack of proper documentation
patients who are refractory to standard therapies or relapse regarding presence of EMM in prospective studies, infor-
after ASCT [16]. Even with ASCT or alloSCT, survival is mation regarding treatment is derived from retrospective
usually not longer than 3 years [15, 38]. Table 3 sum- series. A variety of strategies like that for molecularly
marizes outcome of EMM in various informative studies. In defined high-risk multiple myeloma patients have been
a cumulative analysis that included 1965 patients with used, but a standard therapy has not been established.
multiple myeloma treated with ASCT on TT and non-TT
protocols, those with EMM diagnosed on PET/CT EMM
(excluding EM-B) had significantly inferior OS at 5 years
relative to patients with multiple myeloma (31% vs. 59%, Several case series have reviewed the role of novel therapies
p 0.001) [11]. Similarly, PFS at 5 years was 21% in EMM and ASCT in EMM in a retrospective manner, often
compared with 50% in multiple myeloma (p = 0.001). In a including secondary EMM, which is a terminal event in a
study defining EMM strictly restricted to biopsy-proven previously diagnosed multiple myeloma (Table 3). More-
EM-E, the median OS from the time of extramedullary over, the definitions of EMM are not consistent and the data
diagnosis was 1.3 years [66]. In a prospective study eval- have been collected over long periods with heterogeneous
uating the role of RVD+/− ASCT for frontline therapy, drug combinations and transplant procedures reflecting
EMM (defined as an FDG-avid mass that arose in the soft changing clinical practices and approved therapies of that
tissue and not contiguous to the bone) at diagnosis was an period. Overall, these studies suggest that regimens that
independent prognostic factor for both PFS and OS [74]. contain bortezomib and/ or IMiDs have improved outcome;
In general, the prognosis for EM-E is worse compared however, the gains in PFS and OS are less pronounced
with EM-B. The presence of EMM in the setting of compared with classic multiple myeloma. In a meta-analysis
M. Bhutani et al.

of eight treatment trials (three for transplant eligible and five Daratumumab, an anti-CD38 monoclonal antibody with
for transplant-ineligible patients), patients with EMM (91% direct antimyeloma and immune-modulatory effects, is
EM-B) treated with IMiDs, mainly lenalidomide, or borte- approved for treatment of newly diagnosed and relapsed
zomib had PFS akin to multiple myeloma patients, sug- multiple myeloma. However, there are limited data
gesting that novel agents improved outcome regardless of regarding the efficacy of daratumumab, either alone or in
presence of EMM [62]. Several clinical reports demonstrate combination, in EMM. An updated pooled analysis of phase
effectiveness of bortezomib [78, 79], as well of lenalido- I/II trials that led to accelerated approval of daratumumab
mide and pomalidomide [80, 81]. Thalidomide, on the other monotherapy in heavily pretreated patients reported that
hand, has shown conflicting efficacy. While a report 12% of patients had at least one extramedullary plasmacy-
described three patients with secondary EMM who toma [90]. The overall response rates for patients with and
achieved CR [82], other reports observed poor efficacy of without extramedullary plasmacytomas were 16.7% (n =
this agent [83, 84]. 18) and 33.1% (n = 130), respectively. In a retrospective
Few studies suggest that ASCT can overcome the poor study involving 30 heavily pretreated advanced multiple
prognostic impact of EMM, whereas most studies con- myeloma “real-world” patients, daratumumab-based ther-
sistently show an inferior outcome despite the use of ASCT apy yielded an overall response rate of 36.6% and a median
(Table 3). In a recent retrospective multi-institutional study PFS and OS of 2.3 and 6.6 months, respectively, with
from Europe, a total of 100 patients with EMM received particular poor results in those with EMM (n = 9); only two
ASCT, of which 51.5% had primary EMM [76]. Overall, achieved partial remission, and both progressed shortly (50
ASCT imparted a survival benefit for both EM-E and EM- and 85 days) after treatment initiation [91].
B. Among patients who underwent an ASCT, the median Ongoing front-line randomized trials in transplant-eligible
PFS from diagnosis was 49 months (EM-B: 51.7 months multiple myeloma evaluating the addition of daratumumab
and EM-E: 46.5 months; p = NS) and for those who did (Cassiopea IFM-HOVON study, NCT02541383) or elotuzu-
not receive an ASCT the median PFS was 28.1 months mab (GMMG-HD6 study, NCT02495922) and in transplant-
(p < 0.001). ineligible patients evaluating the addition of daratumumab
Weighing the effect of double versus single ASCT, the (Alcyone study, NCT02195479; Maia study, NCT02252172)
superiority of tandem ASCT cannot be convincingly or elotuzumab (ELOQUENT-1 study, NCT01335399) are
established. In the landmark analyses, tandem ASCT incorporating PET-CT at diagnosis and during patient follow-
showed similar 3-year PFS and OS with HR of 0.83 and up and may therefore inform an optimal strategy for EMM
0.74 (p = 0.13), respectively compared with single ASCT patients.
[63]. Whereas in another study comparing the outcome of
single ASCT (n = 373) versus tandem ASCT (n = 84) in PCL
EMM patients with high-risk cytogenetics (present in 40%),
OS was 70% for single ASCT versus 83% for tandem Table 4 provides a summary of treatment outcome of pri-
ASCT (p = 0.06) and corresponding PFS was 43% vs. 52%, mary PCL with the use of novel agents and transplant in
respectively (p = 0.30) [85]. In multivariable analysis, various informative studies [33, 36, 92–99]. There are two
patients receiving a tandem ASCT were less likely to die prospective phase II studies [33, 92], while other studies are
than were patients who received single ASCT with hazard retrospective. In general, these studies provide evidence for
ratio of 0.46 (0.24–0.89; p = 0.02). the activity of bortezomib-based regimens. In a prospective
The data in EMM with incorporation of carfilzomib, an study, bortezomib, dexamethasone plus doxorubicin or
irreversible second-generation proteasome inhibitor, are not cyclophosphamide induction followed by transplantation
readily available. In a retrospective study investigating (alloSCT, double ASCT, or ASCT-alloSCT) in patients
carfilzomib alone or in combination as salvage therapy in with PCL demonstrated median PFS and OS of 15.1 and
relapsed/refractory multiple myeloma, the presence of 36.3 months, respectively [33]. Among immunomodulatory
EMM resulted in a strong trend towards shorter duration of drugs, the combination of lenalidomide and dexamethasone
response compared with absent EMM (3.9 months vs. was prospectively evaluated in 23 patients with primary
9.3 months, respectively; p = 0.06) [86]. Espanol et al. PCL, 14 patients completed the initial 4 planned cycles with
reported a case with pleuropericardial EMM treated with a achievement of PR in 61%, and ≥VGPR in 35%, 9 patients
combination of carfilzomib and dexamethasone, along with underwent ASCT after induction. After a median follow-up
pericardiocentesis and thoracentesis, resulting in quick CR of 34 months, median PFS was 14 months and median OS
[87]. Based on efficacy of carfilzomib in high molecular risk was 28 months, with superior survival in patients who
patients enrolled on ENDEAVOR study [88] and ASPIRE underwent ASCT than in those who did not [92].
study [89], it can be speculated that carfilzomib may also Registry studies have explored the role of ASCT in PCL.
prove effective in the setting of EMM. The European Group for Blood and Marrow
Table 4 Outcomes for primary PCL in select prospective and retrospective studies after treatment with novel agents and stem cell transplant
Number of primary Treatment plan Median PFS OS Comments Reference
PCL patients (period
covered)

40 (2010–2013) Induction with PAD/VCD. Consolidation 15.1 mo (not reached with double ASCT 36.3 mo (not reached with Prospective phase II study. Royer et al. [33]
with ASCT-alloSCT (n = 17), if matched vs. 17.9 mo with ASCT-alloSCT). double ASCT vs. 36.3 mo with ASCT-alloSCT did not show IFM study group
donor available, otherwise tandem ASCT ASCT-alloSCT). improvement in survival compared with
(n = 7). double ASCT.
Maintenance (for 1 year) with Bz, Dex and
Len in tandem (double) ASCT group.
23 (2009–2011) Induction with Len + Dex. 14 months (27 mo with ASCT vs. 2 mo 28 mo (not reached with ASCT Prospective phase II study. Musto et al. [92]
Extramedullary multiple myeloma

Consolidation with ASCT in eligible without ASCT). vs. 12 mo without ASCT). Survival advantage was exclusively GIMEMA study group
patients. confined to patients who
underwent ASCT
38 (2005–2016) Bz based induction 100%, Bz + IMiD 92%; 20 mo (25 mo with ASCT vs. 6 mo 33 mo (36 mo with ASCT vs. 26 The median PFS was significantly longer Mina et al. [93]
ASCT 74%, and maintenance 61%. without ASCT; 27 mo with ASCT and mo without ASCT) in patients who received ASCT upfront as Single center study
maintenance vs. 11 mo with ASCT (38 mo with ASCT and compared with those who did not.
without maintenance). maintenance vs. 22 mo with The receipt of maintenance therapy after
ASCT without maintenance). ASCT significantly prolonged survival.
Greatest survival benefit was observed
among patients who underwent PI plus
IMiD induction, ASCT, and a 3-drug
maintenance.
23 (2000–2016) PI or IMiDs prior to ASCT 100%; 5.5 mo (18.6 mo with maintenance vs. 18.1 mo (31.8 mo with Gowda et al. [94]
maintenance 52% 3.8 mo without maintenance) maintenance vs. 16.1 mo Single center study
without maintenance)
50 (2000–2016) Novel treatment (mostly Bz based) 80%; 12 mo (18 mo with Bz + ASCT vs. 9 mo 18 mo (48 mo with Bz + ASCT Inclusion of Bz based therapy on the Katodritou et al. [95]
ASCT 40%. in others) vs. 14 mo in others) backbone of ASCT improved OS Greek myeloma
study group
117 (2006–2016) Novel treatment 76%; upfront ASCT 64% NA 23 mo (35 mo with ASCT vs. 13 Better survival with ASCT than without Jurczyszyn et al. [96]
mo without ASCT) International
multicenter study
37 (2002–2016) Bz 77%, IMiDs 67%, cyclophosphamide NA 15 mo (35.5 mo with ASCT) Ganzel et al. [97]
67%, anthracycline 26%, melphalan 13%. Israeli MM study group
SCT 49%- 1/3 of these alloSCT
69 (1998–2015) Novel agents + ASCT 25%, conventional 12.2 mo (26.4 mo with novel agents + 16.1 mo (31.1 mo with novel Jung et al. [98]
chemotherapy + ASCT 12%, novel therapies ASCT vs. 9 mo without ASCT) agents + ASCT vs. 12.3 mo Korean MM
only 36%, conventional chemotherapy without ASCT) working party
only 27%
38 (2001–2012) 64% treated with vincristine, doxorubicin Median PFS not reported 34.2 mo (53.4 mo with Iriuchishima et al. [99]
and dexamethasone, 34% treated with novel maintenance vs. 34.2 mo Japanese
agents +/− ASCT-alloSCT without maintenance Myeloma Society
after ASCT)
27 (not provided) TT1 (n = 7) and TT2 (n = 12) did not 0.8 y vs. 5.4 y (for MM) 1.3 y vs. 8.8 y for MM Usmani et al. [36]
include novel agents. TT3 (n = 8) included Single Center Study
novel agents.

PCL plasma cell leukemia, PFS progression-free survival, OS overall survival, PAD (Bortezomib, Doxorubicin, Dexamethasone), VCD (Bortezomib, Cyclophosphamide, Dexamethasone), ASCT
autologous stem cell transplant, alloSCT allogeneic stem cell transplant, Bz bortezomib, Len lenaldiomide, Dex dexamethasone, IMiD immunomodulatory drug, PI proteasome inhibitor, TT total
therapy, y years, mo months, MM multiple myeloma, NA not available
M. Bhutani et al.

Transplantation registry reported the outcomes of 272 therefore merit investigation for treatment of EMM and
patients with primary PCL who underwent an ASCT PCL, often harboring high-risk molecular abnormalities.
between 1980 and 2006 [100], quoting a median PFS and Emerging cellular therapies including CAR-T cell therapy
OS of 14.3 months and 25.7 months, respectively. Simi- also hold promise to improve the prognosis, therefore it
larly, the Center for International Blood and Marrow would be desirable to enroll patients with EMM and PCL in
Transplant Research (CIBMTR) showed a survival benefit prospective studies designed for multiple myeloma.
with upfront ASCT for 97 patients with primary PCL
treated between 1995 and 2006 [101]. The 3-year PFS and
OS were 34 and 64%, respectively. In addition, there Management approach
appeared to be a trend toward a superior OS in patients who
received a tandem ASCT as opposed to single ASCT, with PET/CT imaging plays an integral role in diagnosis and
OS at 3 years being 84 and 56%, respectively. Since the monitoring of treatment response at various extramedullary
analyzed cohorts represent a selected group of patients that sites [7]. Biopsy of EMM lesion must be considered for
had survived long enough to undergo transplantation and acquisition of molecular information, and to establish
had demonstrated chemotherapy-sensitive disease, the diagnosis in nonsecretory cases with no marrow involve-
observed outcome is likely expected to be better than for an ment. The main goal of therapy is to rapidly debulk the
unselected group of patients with EMM and PCL. In a more disease in both bone marrow and extramedullary sites and
recent study, patients with primary PCL who received an to reverse end-organ complications, to prevent early mor-
intensive strategy including proteasome inhibitor plus IMiD tality and eventually prolong survival.
induction regimen, consolidation with a single ASCT, and The current treatment approach is tailored to patient age
followed by a 3‐drug maintenance regimen, the median PFS and fitness, with fit patients receiving intensive induction
was 33 months, and the median OS was 63 months, with 3‐ therapy, consolidation ASCT and maintenance, whereas
year PFS and OS rates of 47 and 58%, respectively [93]. unfit patients are treated with several cycles of combination
The role of alloSCT has been described in small retro- chemotherapy and extended maintenance. Radiation ther-
spective studies. In the CIBMTR series, the outcome of 97 apy is employed in selected cases with bulky disease to
patients who received ASCT was compared with 50 patients improve local control and palliate symptoms. Where pos-
who received alloSCT between 1995 and 2006. Although sible, patients should be considered for enrollment in clin-
the cumulative incidence of relapse at 3 years was sig- ical trials.
nificantly lower in the allogeneic group (38% vs. 61%), In younger/fit patients with proliferative/ bulky EMM or
transplant-related mortality at 3 years was considerably PCL, more intensive combination regimens incorporating a
higher in patients who received an alloSCT (41% vs. 5%) proteasome inhibitor and alkylators, such as V(or K)RD-
versus ASCT. This resulted in a 3-year OS of 64% for PACE (bortezomib or carfilzomib, lenalidomide, dex-
ASCT group and 39% for the alloSCT group, respectively. amethasone, cisplatin, doxorubicin, cyclophosphamide, and
While a comparison between ASCT and alloSCT is difficult etoposide) may be utilized to provide rapid reduction in
owing to relatively small numbers of alloSCT and lack of disease burden (Fig. 2). Echocardiographic estimation of
randomized studies, the available evidence does not suggest cardiac function is recommended prior to starting therapy.
that alloSCT is superior to ASCT, despite decreased rates of Treating EMM is analogous to treating aggressive lym-
disease relapse. phomas or acute leukemia, with propensity for tumor cells
The European Myeloma Network study (EMN12/ to rapidly grow and develop drug resistance during inter-
HOVON 129 PCL) is analyzing the role of carfilzomib and current periods of suboptimal treatment, prolonged treat-
lenalidomide containing induction regimen, followed by ment breaks or delays. Avoiding low-intensity suboptimal
tandem ASCT and alloSCT, the latter involving semi- treatment is the crux. Patients must receive tumor lysis
intensive conditioning with melphalan 140 mg/m2 + flu- prophylaxis with hydration and allopurinol. In patients with
darabine, as well as carfilzomib and lenalidomide in con- underlying renal disease, and/or history of allopurinol
solidation and maintenance in primary PCL. Venetoclax intolerance, the use of febuxostat (a new xanthine oxidase
(BCL2 inhibitor) has shown significant activity in patients inhibitor) may be considered. Rasburicase is preferred in
with t(11;14) + multiple myeloma, a translocation fre- patients whose baseline uric acid is higher than 8 mg/dL
quently reported in primary PCL, thus providing the ratio- (476 mmol/L) or those who develop evidence of active
nale for exploring this drug in PCL in combination with tumor lysis. Intensive supportive care is critical with careful
other agents. Monoclonal antibodies directed against CD38 monitoring of cytopenia and nearly all patients will require
and SLAMF7, exportin 1 inhibitors (e.g., selinexor), anti- blood product support. Febrile neutropenia is a frequent
BCMA antibodies, and bispecific T-cell engagers have complication, particularly after PACE based therapies, and
shown efficacy in high-risk multiple myeloma, and patients must be counseled to seek immediate medical
Extramedullary multiple myeloma

Fig. 2 Management approach


for EMM or primary PCL.
**Consider for patients who are
fit enough to receive aggressive
chemotherapy. In the absence of
PR or better, approach as
primary refractory and switch to
2nd line regimen. *Consider
reduced-intensity allogeneic
stem cell transplant for young/fit
patients in the context of a
clinical trial only. PCL plasma
cell leukemia, EMM
Extramedullary multiple
myeloma, RVd lenalidomide,
bortezomib, dexamethasone,
KRd carfilzomib, lenalidomide,
dexamethasone, V(K)-RD-
PACE bortezomb(carfilzomib)-
lenalidomide, dexamethasone,
cisplatin, doxorubicin,
cyclophosphamide, etoposide,
PR partial response, CR
complete response, CSF
cerebrospinal fluid, CNS central
nervous system

attention with fever. Antibiotic prophylaxis directed against After induction therapy, in transplant-eligible patients, an
gram-negative bacteria, and antifungal prophylaxis should upfront ASCT is recommended to achieve a deeper
be considered if neutropenia is anticipated for a long response and likely longer disease control. Based on limited
duration. Adequate antithrombotic prophylaxis is also cru- data showing efficacy, a tandem ASCT could be considered
cial. Finally, for all patients with EMM, we recommend the [63, 101]. An alloSCT may be considered and performed
use bisphosphonate therapy or denosumab (for patients with preferably only in the setting of a clinical trial. In the
renal impairment) to decrease the risk of future skeletal- posttransplant period, our group minimizes the period with
related events. no therapy to 60 days rather than the conventional 100 days
In cases with less bulky or low-volume EMM lacking the for the initiation of consolidation/maintenance therapy, as is
features of PCL, or for those with pre-existing organ dys- done in the total therapy program from Arkansas. The
function, or significant comorbidities, we prefer frontline maintenance regimens typically include a proteasome
therapy with a triplet regimen that includes a proteasome inhibitor and an IMiD combination, given promising results
inhibitor and an IMiD, such as RVd (lenalidomide, borte- with this strategy in high-risk multiple myeloma [102]. The
zomib, and dexamethasone) or KRd (carfilzomib, lenali- approach of prolonged administration of RVD-based
domide, and dexamethasone). In the very elderly population maintenance forms the basis for the current ongoing
and in frail patients, treatment efficacy should be carefully Southwest Oncology Group clinical trial for high risk as the
weighed against the risk of life-threatening adverse events control arm (elotuzumab plus RVD vs. RVD for high risk
that may significantly impair quality of life. In these situa- only with maintenance until progression in each arm;
tions, dose reductions could be adopted, or in selected cases SWOG S1211, NCT01668719).
a palliative approach could be offered upfront.
Patients failing to achieve a partial response after two
cycles of induction therapy or breaking through the treat- CNS EMM
ment, if remain eligible, should be timely switched to a
more aggressive regimen to minimize risk of organ failure The CNS is an immunologically privileged site that is iso-
and death. lated from the blood system by the blood–brain barrier
M. Bhutani et al.

(BBB) and the blood–cerebrospinal fluid (CSF) barriers. trials. It is unknown whether these antibodies can get
Although the process by which lymphoid or myeloid leu- through the blood–brain barrier or be used intrathecally.
kemic cells traffic into the CNS has been studied, this Engineered CAR-T cells targeting BCMA antigens were
process is less well understood in the context of malignant found in CSF of several patients recruited to dedicated
plasma cells. CNS is an exceedingly rare location of EMM trials, suggesting these might have a role in eradicating CNS
involvement, diagnosed at an incidence of less than disease. Our approach for CNS EMM includes IT che-
1%, with an overall survival reported less than 6 months motherapy, CNS radiotherapy in selected cases, and sys-
[103–105]. Most patients are diagnosed with CNS invol- temic IMiD-based combination therapy.
vement at relapse/progression. Diagnosis is made by pre-
sence of atypical plasma cells in the CSF by conventional
cytology and flowcytometry or direct tissue sampling and/or Conclusions
imaging evidence of intraparenchymal lesions or leptome-
ningeal/dural enhancement. A correct and timely diagnosis Despite greatly improved prognosis for multiple myeloma
still represents a challenge as neurological symptoms and in general, our current standard therapies have not suffi-
signs may be subtle, and sometimes attributed to other ciently improved outcomes for patients with EMM. The
clinical situations, such as hyperviscosity, metabolic ence- rarity of this disease and the lack of prospective trials make
phalopathy from hypercalcemia or AKI, treatment-related it difficult to generate solid data and treatment guidelines.
neuropathy, stroke or opportunistic infections. The fact that Although it will be challenging to have trials adequately
many patients with CNS EMM have PCL or other extra- powered to investigate outcomes in this uncommon group
medullary localizations suggests that the hematologic route of patients, trials targeting ‘high-risk multiple myeloma’
plays a major role for spreading [106]. Besides hemato- could allow inclusion of these patients for assessment of
genous spread, CNS EMM can arise from direct invasion reported subgroup effect estimates with a priori hypotheses.
from skull or vertebral lesions into the CNS. MRI with Capturing information regarding EM-B and EM-E at diag-
gadolinium contrast has a superior sensitivity than cranial nosis and at the time of relapse in clinical trials incorpor-
CT to assess for CNS EMM, whereas PET/CT is best suited ating sensitive imaging modalities will be a key to
to localize involvement of sites outside the CNS. evaluating outcomes for EMM prospectively. International
Management of CNS EMM represents a unique chal- collaborative studies are warranted to better understand the
lenge. Most published reports describe dismal survival with risk factors, the biological and genetic features and the
traditional use of intrathecal chemotherapy and radiation impact of novel therapeutic modalities. As more data are
therapy with or without systemic therapy [106, 107]. In the accumulated from clinical trials evaluating next wave of
largest retrospective multi-institutional study of 172 CNS antimyeloma therapies, including selinexor, checkpoint
EMM cases, the median OS from the onset of CNS invol- inhibitors, CAR T cells, antibody-drug conjugates and
vement for the entire group was 7 months; untreated and bispecific antibodies, a better understanding of how to
treated patients had median OS of 2 and 8 months, sequence and cycle therapies will be critical to further
respectively [104]. Despite the use of novel agents, or improving outcomes in EMM.
aggressive therapies, in combination with ASCT or
alloSCT, the outcomes after CNS EMM diagnosis are Acknowledgements This work is supported by the Carolinas Mye-
loma Research Fund, Heinemann Foundation of Charlotte, the
uniformly dismal [103, 108, 109].
Freedland Fund, the Leukemia Lymphoma Society and NCI Grant
Alkylating agents penetrate the CSF poorly, and con- 5R01CA201634.
ventional agents like high-dose methotrexate and cytar-
abine, known to cross the BBB, have minimal to no Author contributions SZU and MB conceived and designed the study.
antimyeloma activity. IMiDs, including thalidomide, lena- MB wrote the first draft of the manuscript with input from SZU and
PMV. DMF, MB and SA prepared the figures. All authors were
lidomide, and pomalidomide have shown CNS penetrance involved in researching data, discussion of content and critical revision
and effectiveness in clearance of myeloma cells from CSF of the paper and approval of the submitted manuscript.
[110, 111]; however, IMiD resistance is not uncommon in
this group of patients. Although proteasome inhibitors Compliance with ethical standards
(bortezomib, carfilzomib, and ixazomib) cannot cross the
BBB, marizomib, an irreversible proteasome inhibitor has Conflict of interest MB has received speaker’s fees from Amgen.
shown to distribute uniformly within the brain parenchyma, DMF has no conflict of interest. SA has received consulting fees from
Takeda, Amgen and Celgene, and speaker’s fees from Takeda and
thus making it a suitable agent to be tested in clinical trials
Celgene. PMV has received consulting fees from Amgen, BMS,
[112]. The anti-CD38 antibody daratumumab and anti-CS1 Celgene, Janssen, Novartis, Oncopeptides, Takeda and TeneoBio.
elotuzumab have not been tested in CNS EMM as CNS SZU has received consulting fees from Abbvie, Amgen, BMS, Cel-
involvement was an exclusion criterion in the registration gene, EdoPharma, GSK, Janssen, Sanofi, Seattle Genetics, Skyline Dx,
Extramedullary multiple myeloma

Takeda and TeneoBio, and research funding from Amgen, Array 16. Pour L, Sevcikova S, Greslikova H, Kupska R, Majkova P,
Biopharma, BMS, Celgene, Janssen, Pharmacyclics, Prothena, Sanofi, Zahradova L, et al. Soft-tissue extramedullary multiple myeloma
Seattle Genetics, Skyline Dx and Takeda. prognosis is significantly worse in comparison to bone-related
extramedullary relapse. Haematologica. 2014;99:360–4.
Publisher’s note Springer Nature remains neutral with regard to 17. Batsukh K, Lee SE, Min GJ, Park SS, Jeon YW, Yoon JH, et al.
jurisdictional claims in published maps and institutional affiliations. Distinct clinical outcomes between paramedullary and extra-
medullary lesions in newly diagnosed multiple myeloma.
Immune Netw. 2017;17:250–60.
18. Dawson MA, Patil S, Spencer A. Extramedullary relapse of
References multiple myeloma associated with a shift in secretion from intact
immunoglobulin to light chains. Haematologica. 2007;92:143–4.
1. Rajkumar SV, Dimopoulos MA, Palumbo A, Blade J, Merlini G, 19. Creach KM, Foote RL, Neben-Wittich MA, Kyle RA. Radio-
Mateos MV, et al. International Myeloma Working Group therapy for extramedullary plasmacytoma of the head and neck.
updated criteria for the diagnosis of multiple myeloma. Lancet Int J Radiat Oncol Biol Phys. 2009;73:789–94.
Oncol. 2014;15:e538–48. 20. Dimopoulos MA, Hamilos G. Solitary bone plasmacytoma and
2. Blade J, Fernandez de Larrea C, Rosinol L, Cibeira MT, Jimenez extramedullary plasmacytoma. Curr Treat Options Oncol. 2002;
R, Powles R. Soft-tissue plasmacytomas in multiple myeloma: 3:255–9.
incidence, mechanisms of extramedullary spread, and treatment 21. Ravi P, Kumar SK, Roeker L, Gonsalves W, Buadi F, Lacy MQ,
approach. J Clin Oncol. 2011;29:3805–12. et al. Revised diagnostic criteria for plasma cell leukemia: results
3. Vande Broek I, Vanderkerken K, Van Camp B, Van Riet I. of a Mayo Clinic study with comparison of outcomes to multiple
Extravasation and homing mechanisms in multiple myeloma. myeloma. Blood Cancer J. 2018;8:116.
Clin Exp Metastasis. 2008;25:325–34. 22. Tiedemann RE, Gonzalez-Paz N, Kyle RA, Santana-Davila R,
4. Weinstock M, Ghobrial IM. Extramedullary multiple myeloma. Price-Troska T, Van Wier SA, et al. Genetic aberrations and
Leuk Lymphoma. 2013;54:1135–41. survival in plasma cell leukemia. Leukemia. 2008;22:1044–52.
5. Fernandez de Larrea C, Kyle RA, Durie BG, Ludwig H, Usmani 23. Garcia-Sanz R, Orfao A, Gonzalez M, Tabernero MD, Blade J,
S, Vesole DH, et al. Plasma cell leukemia: consensus statement Moro MJ, et al. Primary plasma cell leukemia: clinical, immu-
on diagnostic requirements, response criteria and treatment nophenotypic, DNA ploidy, and cytogenetic characteristics.
recommendations by the International Myeloma Working Group. Blood. 1999;93:1032–7.
Leukemia. 2013;27:780–91. 24. Morgan GJ, Walker BA, Davies FE. The genetic architecture of
6. International Myeloma Working Group. Criteria for the classi- multiple myeloma. Nat Rev Cancer. 2012;12:335–48.
fication of monoclonal gammopathies, multiple myeloma and 25. Rasche L, Chavan SS, Stephens OW, Patel PH, Tytarenko R,
related disorders: a report of the International Myeloma Working Ashby C, et al. Spatial genomic heterogeneity in multiple mye-
Group. Br J Haematol. 2003;121:749–57. loma revealed by multi-region sequencing. Nat Commun.
7. Touzeau C, Moreau P. How I treat extramedullary myeloma. 2017;8:268.
Blood. 2016;127:971–6. 26. Pawlyn C, Morgan GJ. Evolutionary biology of high-risk mul-
8. Rasche L, Bernard C, Topp MS, Kapp M, Duell J, Wesemeier C, tiple myeloma. Nat Rev Cancer. 2017;17:543–56.
et al. Features of extramedullary myeloma relapse: high pro- 27. Bergsagel PL, Kuehl WM, Zhan F, Sawyer J, Barlogie B,
liferation, minimal marrow involvement, adverse cytogenetics: a Shaughnessy J Jr. Cyclin D dysregulation: an early and uni-
retrospective single-center study of 24 cases. Ann Hematol. fying pathogenic event in multiple myeloma. Blood. 2005;
2012;91:1031–7. 106:296–303.
9. Chang H, Qi X, Yeung J, Reece D, Xu W, Patterson B. Genetic 28. Keats JJ, Maxwell CA, Taylor BJ, Hendzel MJ, Chesi M,
aberrations including chromosome 1 abnormalities and clinical Bergsagel PL, et al. Overexpression of transcripts originating
features of plasma cell leukemia. Leuk Res. 2009;33:259–62. from the MMSET locus characterizes all t(4;14)(p16;q32)-posi-
10. Cifola I, Lionetti M, Pinatel E, Todoerti K, Mangano E, Pietrelli tive multiple myeloma patients. Blood. 2005;105:4060–9.
A, et al. Whole-exome sequencing of primary plasma cell leu- 29. Jelinek T, Bezdekova R, Zatopkova M, Burgos L, Simicek M,
kemia discloses heterogeneous mutational patterns. Oncotarget. Sevcikova T, et al. Current applications of multiparameter flow
2015;6:17543–58. cytometry in plasma cell disorders. Blood Cancer J. 2017;7:e617.
11. Usmani SZ, Heuck C, Mitchell A, Szymonifka J, Nair B, 30. Jelinek T, Kryukov F, Rihova L, Hajek R. Plasma cell leukemia:
Hoering A, et al. Extramedullary disease portends poor prognosis from biology to treatment. Eur J Haematol. 2015;95:16–26.
in multiple myeloma and is over-represented in high-risk 31. Rihova L, Vsianska P, Bezdekova R, Adam Z, Penka M, Jelinek
disease even in the era of novel agents. Haematologica. 2012; T, et al. Identification of phenotype profile related to the extra-
97:1761–7. medullary involvement in multiple myeloma relapse. Blood.
12. Pasmantier MW, Azar HA. Extraskeletal spread in multiple 2016;128:5653–5653.
plasma cell myeloma. A review of 57 autopsied cases. Cancer. 32. Deng S, Xu Y, An G, Sui W, Zou D, Zhao Y, et al. Features of
1969;23:167–74. extramedullary disease of multiple myeloma: high frequency of
13. Blade J, Lust JA, Kyle RA. Immunoglobulin D multiple mye- p53 deletion and poor survival: a retrospective single-center
loma: presenting features, response to therapy, and survival in a study of 834 cases. Clin Lymphoma Myeloma Leuk. 2015;
series of 53 cases. J Clin Oncol. 1994;12:2398–404. 15:286–91.
14. Blade J, Kyle RA, Greipp PR. Presenting features and prognosis 33. Royer B, Minvielle S, Diouf M, Roussel M, Karlin L, Hulin C,
in 72 patients with multiple myeloma who were younger than 40 et al. Bortezomib, doxorubicin, cyclophosphamide, dex-
years. Br J Haematol. 1996;93:345–51. amethasone induction followed by stem cell transplantation for
15. Varettoni M, Corso A, Pica G, Mangiacavalli S, Pascutto C, primary plasma cell leukemia: a prospective phase II study of the
Lazzarino M. Incidence, presenting features and outcome of intergroupe francophone du myelome. J Clin Oncol. 2016;34:
extramedullary disease in multiple myeloma: a longitudinal 2125–32.
study on 1003 consecutive patients. Ann Oncol. 2010;21: 34. Avet-Loiseau H, Malard F, Campion L, Magrangeas F, Sebban
325–30. C, Lioure B, et al. Translocation t(14;16) and multiple myeloma:
M. Bhutani et al.

is it really an independent prognostic factor? Blood. 2011; 52. Chang H, Bartlett ES, Patterson B, Chen CI, Yi QL. The absence
117:2009–11. of CD56 on malignant plasma cells in the cerebrospinal fluid is
35. Neri A, Todoerti K, Lionetti M, Simeon V, Barbieri M, Nozza F, the hallmark of multiple myeloma involving central nervous
et al. Primary plasma cell leukemia 2.0: advances in biology and system. Br J Haematol. 2005;129:539–41.
clinical management. Expert Rev Hematol. 2016;9:1063–73. 53. Liebisch P, Eppinger S, Schopflin C, Stehle G, Munzert G,
36. Usmani SZ, Nair B, Qu P, Hansen E, Zhang Q, Petty N, et al. Dohner H, et al. CD44v6, a target for novel antibody treatment
Primary plasma cell leukemia: clinical and laboratory presenta- approaches, is frequently expressed in multiple myeloma and
tion, gene-expression profiling and clinical outcome with Total associated with deletion of chromosome arm 13q. Haematolo-
Therapy protocols. Leukemia. 2012;26:2398–405. gica. 2005;90:489–93.
37. de Haart SJ, Willems SM, Mutis T, Koudijs MJ, van Blokland 54. van Driel M, Gunthert U, Stauder R, Joling P, Lokhorst HM,
MT, Lokhorst HM, et al. Comparison of intramedullary mye- Bloem AC. CD44 isoforms distinguish between bone marrow
loma and corresponding extramedullary soft tissue plasmacyto- plasma cells from normal individuals and patients with
mas using genetic mutational panel analyses. Blood Cancer J. multiple myeloma at different stages of disease. Leukemia.
2016;6:e426. 1998;12:1821–8.
38. Varga C, Xie W, Laubach J, Ghobrial IM, O’Donnell EK, 55. Azab AK, Quang P, Azab F, Pitsillides C, Thompson B,
Weinstock M, et al. Development of extramedullary myeloma Chonghaile T, et al. P-selectin glycoprotein ligand regulates the
in the era of novel agents: no evidence of increased risk interaction of multiple myeloma cells with the bone marrow
with lenalidomide-bortezomib combinations. Br J Haematol. microenvironment. Blood. 2012;119:1468–78.
2015;169:843–50. 56. Hargreaves DC, Hyman PL, Lu TT, Ngo VN, Bidgol A, Suzuki
39. Lopez-Anglada L, Gutierrez NC, Garcia JL, Mateos MV, Flores G, et al. A coordinated change in chemokine responsiveness
T, San Miguel JF. P53 deletion may drive the clinical evolution guides plasma cell movements. J Exp Med. 2001;194:45–56.
and treatment response in multiple myeloma. Eur J Haematol. 57. Roccaro AM, Mishima Y, Sacco A, Moschetta M, Tai YT, Shi J,
2010;84:359–61. et al. CXCR4 regulates extra-medullary myeloma through
40. Chang H, Sloan S, Li D, Keith Stewart A. Multiple myeloma epithelial-mesenchymal-transition-like transcriptional activation.
involving central nervous system: high frequency of chromo- Cell Rep. 2015;12:622–35.
some 17p13.1 (p53) deletions. Br J Haematol. 2004;127:280–4. 58. Azab AK, Hu J, Quang P, Azab F, Pitsillides C, Awwad R, et al.
41. Sheth N, Yeung J, Chang H. p53 nuclear accumulation is asso- Hypoxia promotes dissemination of multiple myeloma through
ciated with extramedullary progression of multiple myeloma. acquisition of epithelial to mesenchymal transition-like features.
Leuk Res. 2009;33:1357–60. Blood. 2012;119:5782–94.
42. Billecke L, Murga Penas EM, May AM, Engelhardt M, Nagler 59. Wang SY, Hao HL, Deng K, Li Y, Cheng ZY, Lv C, et al.
A, Leiba M, et al. Cytogenetics of extramedullary manifestations Expression levels of phosphatase and tensin homolog deleted
in multiple myeloma. Br J Haematol. 2013;161:87–94. on chromosome 10 (PTEN) and focal adhesion kinase in
43. Bezieau S, Devilder MC, Avet-Loiseau H, Mellerin MP, Puthier patients with multiple myeloma and their relationship to clin-
D, Pennarun E, et al. High incidence of N and K-Ras activating ical stage and extramedullary infiltration. Leuk Lymphoma.
mutations in multiple myeloma and primary plasma cell leuke- 2012;53:1162–8.
mia at diagnosis. Hum Mutat. 2001;18:212–24. 60. Vacca A, Di Loreto M, Ribatti D, Di Stefano R, Gadaleta-
44. Mulligan G, Lichter DI, Di Bacco A, Blakemore SJ, Berger A, Caldarola G, Iodice G, et al. Bone marrow of patients with active
Koenig E, et al. Mutation of NRAS but not KRAS significantly multiple myeloma: angiogenesis and plasma cell adhesion
reduces myeloma sensitivity to single-agent bortezomib therapy. molecules LFA-1, VLA-4, LAM-1, and CD44. Am J Hematol.
Blood. 2014;123:632–9. 1995;50:9–14.
45. Egan JB, Kortuem KM, Kurdoglu A, Izatt T, Aldrich J, Reiman 61. Hedvat CV, Comenzo RL, Teruya-Feldstein J, Olshen AB,
R, et al. Extramedullary myeloma whole genome sequencing Ely SA, Osman K, et al. Insights into extramedullary tumour
reveals novel mutations in Cereblon, proteasome subunit G2 and cell growth revealed by expression profiling of human plas-
the glucocorticoid receptor in multi drug resistant disease. Br J macytomas and multiple myeloma. Br J Haematol. 2003;
Haematol. 2013;161:748–51. 122:728–44.
46. Schinke CD, Ashby C, Wang Y, Tytarenko RG, Boyle E, 62. Montefusco V, Gay F, Spada S, De Paoli L, Di Raimondo F,
Wardell C, et al. The mutational landscape of primary plasma Ribolla R, et al. Outcome of paraosseous extra-medullary disease
cell leukemia. Blood. 2018;132:114–114. in newly diagnosed multiple myeloma patients treated with new
47. Van Riet I, Vanderkerken K, de Greef C, Van Camp B. Homing drugs. Haematologica. 2019; pii: haematol. 2019.219139. https://
behaviour of the malignant cell clone in multiple myeloma. Med doi.org/10.3324/haematol.2019.219139.
Oncol. 1998;15:154–64. 63. Gagelmann N, Eikema DJ, Iacobelli S, Koster L, Nahi H,
48. Vanderkerken K, Van Camp B, De Greef C, Vande Broek I, Stoppa AM, et al. Impact of extramedullary disease in patients
Asosingh K, Van Riet I. Homing of the myeloma cell clone. Acta with newly diagnosed multiple myeloma undergoing auto-
Oncol. 2000;39:771–6. logous stem cell transplantation: a study from the Chronic
49. Moschetta M, Kawano Y, Sacco A, Belotti A, Ribolla R, Malignancies Working Party of the EBMT. Haematologica.
Chiarini M, et al. Bone marrow stroma and vascular contribu- 2018;103:890–7.
tions to myeloma bone homing. Curr Osteoporos Rep. 2017;15: 64. Mangiacavalli S, Pompa A, Ferretti V, Klersy C, Cocito F,
499–506. Varettoni M, et al. The possible role of burden of therapy on the
50. Pellat-Deceunynck C, Barille S, Puthier D, Rapp MJ, Har- risk of myeloma extramedullary spread. Ann Hematol. 2017;
ousseau JL, Bataille R, et al. Adhesion molecules on human 96:73–80.
myeloma cells: significant changes in expression related to 65. Kumar L, Gogi R, Patel AK, Mookerjee A, Sahoo RK, Malik PS,
malignancy, tumor spreading, and immortalization. Cancer Res. et al. Multiple myeloma with extramedullary disease: impact of
1995;55:3647–53. autologous stem cell transplantation on outcome. Bone Marrow
51. Dahl IM, Rasmussen T, Kauric G, Husebekk A. Differential Transpl. 2017;52:1473–5.
expression of CD56 and CD44 in the evolution of extra- 66. Weinstock M, Aljawai Y, Morgan EA, Laubach J, Gannon M,
medullary myeloma. Br J Haematol. 2002;116:273–7. Roccaro AM, et al. Incidence and clinical features of
Extramedullary multiple myeloma

extramedullary multiple myeloma in patients who underwent in patients with multiple myeloma: report of 2 cases. Clin
stem cell transplantation. Br J Haematol. 2015;169:851–8. Lymphoma Myeloma Leuk. 2013;13:83–5.
67. Short KD, Rajkumar SV, Larson D, Buadi F, Hayman S, Dis- 81. Nakazato T, Mihara A, Ito C, Sanada Y, Aisa Y. Lenalidomide is
penzieri A, et al. Incidence of extramedullary disease in patients active for extramedullary disease in refractory multiple myeloma.
with multiple myeloma in the era of novel therapy, and the Ann Hematol. 2012;91:473–4.
activity of pomalidomide on extramedullary myeloma. Leuke- 82. Biagi JJ, Prince HM. Thalidomide is effective for extramedullary
mia. 2011;25:906–8. relapse of multiple myeloma post-allogeneic bone marrow
68. Minnema MC, van de Donk NW, Zweegman S, Hegenbart U, transplantation. Br J Haematol. 2001;115:484–5.
Schonland S, Raymakers R, et al. Extramedullary relapses after 83. Blade J, Perales M, Rosinol L, Tuset M, Montoto S, Esteve J,
allogeneic non-myeloablative stem cell transplantation in multi- et al. Thalidomide in multiple myeloma: lack of response of soft-
ple myeloma patients do not negatively affect treatment outcome. tissue plasmacytomas. Br J Haematol. 2001;113:422–4.
Bone Marrow Transpl. 2008;41:779–84. 84. Rosinol L, Cibeira MT, Blade J, Esteve J, Aymerich M, Rozman
69. Perez-Simon JA, Sureda A, Fernandez-Aviles F, Sampol A, M, et al. Extramedullary multiple myeloma escapes the effect of
Cabrera JR, Caballero D, et al. Reduced-intensity conditioning thalidomide. Haematologica. 2004;89:832–6.
allogeneic transplantation is associated with a high incidence of 85. Gagelmann N, Eikema DJ, Koster L, Caillot D, Pioltelli P,
extramedullary relapses in multiple myeloma patients. Leukemia. Lleonart JB, et al. Tandem autologous stem cell transplantation
2006;20:542–5. improves outcome in newly diagnosed multiple myeloma with
70. Raanani P, Shpilberg O, Ben-Bassat I. Extramedullary disease extramedullary disease and high-risk cytogenetics: a study from
and targeted therapies for hematological malignancies-is the the Chronic Malignancies Working Party of EBMT. Biol Blood
association real? Ann Oncol. 2007;18:7–12. Marrow Transplant. 2019;25:2134–42.
71. Ali R, Ozkalemkas F, Ozkan A, Ozkocaman V, Ozcelik T, Ozan 86. Muchtar E, Gatt ME, Rouvio O, Ganzel C, Chubar E, Suriu C,
U, et al. Bortezomib and extramedullary disease in multiple et al. Efficacy and safety of salvage therapy using Carfilzomib
myeloma: the shine and dark side of the moon. Leuk Res. for relapsed or refractory multiple myeloma patients: a multi-
2007;31:1153–5. centre retrospective observational study. Br J Haematol. 2016;
72. Zeiser R, Deschler B, Bertz H, Finke J, Engelhardt M. Extra- 172:89–96.
medullary vs medullary relapse after autologous or allogeneic 87. Espanol I, Romera M, Gutierrez-Meca MD, Garcia MDC,
hematopoietic stem cell transplantation (HSCT) in multiple Tejedor A, Martinez A, et al. Carfilzomib and dexamethasone for
myeloma (MM) and its correlation to clinical outcome. Bone extramedullary myeloma with pleuropericardial involvement.
Marrow Transpl. 2004;34:1057–65. Clin Case Rep. 2017;5:1258–60.
73. Alegre A, Granda A, Martinez-Chamorro C, Diaz-Mediavilla J, 88. Chng WJ, Goldschmidt H, Dimopoulos MA, Moreau P, Joshua
Martinez R, Garcia-Larana J, et al. Different patterns of relapse D, Palumbo A, et al. Carfilzomib-dexamethasone vs bortezomib-
after autologous peripheral blood stem cell transplantation in dexamethasone in relapsed or refractory multiple myeloma by
multiple myeloma: clinical results of 280 cases from the Spanish cytogenetic risk in the phase 3 study ENDEAVOR. Leukemia.
Registry. Haematologica. 2002;87:609–14. 2017;31:1368–74.
74. Moreau P, Attal M, Caillot D, Macro M, Karlin L, Garderet L, 89. Dimopoulos M, Wang M, Maisnar V, Minarik J, Bensinger W,
et al. Prospective evaluation of magnetic resonance imaging and Mateos MV, et al. Response and progression-free survival
[(18)f]fluorodeoxyglucose positron emission tomography- according to planned treatment duration in patients with relapsed
computed tomography at diagnosis and before maintenance multiple myeloma treated with carfilzomib, lenalidomide, and
therapy in symptomatic patients with multiple myeloma included dexamethasone (KRd) versus lenalidomide and dexamethasone
in the IFM/DFCI 2009 trial: results of the IMAJEM study. J Clin (Rd) in the phase III ASPIRE study. J Hematol Oncol. 2018;
Oncol. 2017;35:2911–8. 11:49.
75. Cavo M, Terpos E, Nanni C, Moreau P, Lentzsch S, Zweegman 90. Usmani SZ, Weiss BM, Plesner T, Bahlis NJ, Belch A, Lonial S,
S, et al. Role of (18)F-FDG PET/CT in the diagnosis and man- et al. Clinical efficacy of daratumumab monotherapy in patients
agement of multiple myeloma and other plasma cell disorders: a with heavily pretreated relapsed or refractory multiple myeloma.
consensus statement by the International Myeloma Working Blood. 2016;128:37–44.
Group. Lancet Oncol. 2017;18:e206–e217. 91. Pick M, Vainstein V, Goldschmidt N, Lavie D, Libster D,
76. Beksac M, Cengiz Seval G, Kanellias N, Coriu D, Rosinol L, Gural A, et al. Daratumumab resistance is frequent in advanced-
Ozet G, et al. A real world multicenter retrospective study on stage multiple myeloma patients irrespective of CD38 expres-
extramedullary disease from Balkan Myeloma Study Group and sion and is related to dismal prognosis. Eur J Haematol.
Barcelona University: analysis of parameters that improve out- 2018;100:494–501.
come. Haematologica. 2019; pii: haematol. 2019.219295. https:// 92. Musto P, Simeon V, Martorelli MC, Petrucci MT, Cascavilla N,
doi.org/10.3324/haematol.2019.219295. [Epub ahead of print]. Di Raimondo F, et al. Lenalidomide and low-dose dex-
77. Gonsalves WI, Rajkumar SV, Go RS, Dispenzieri A, Gupta V, amethasone for newly diagnosed primary plasma cell leukemia.
Singh PP, et al. Trends in survival of patients with primary Leukemia. 2014;28:222–5.
plasma cell leukemia: a population-based analysis. Blood. 93. Mina R, Joseph NS, Kaufman JL, Gupta VA, Heffner LT,
2014;124:907–12. Hofmeister CC, et al. Survival outcomes of patients with primary
78. Laura R, Cibeira MT, Uriburu C, Yantorno S, Salamero O, Blade plasma cell leukemia (pPCL) treated with novel agents. Cancer.
J, et al. Bortezomib: an effective agent in extramedullary disease 2019;125:416–23.
in multiple myeloma. Eur J Haematol. 2006;76:405–8. 94. Gowda L, Shah M, Badar I, Bashir Q, Shah N, Patel K, et al.
79. Patriarca F, Prosdocimo S, Tomadini V, Vasciaveo A, Bruno B, Primary plasma cell leukemia: autologous stem cell transplant in
Fanin R. Efficacy of bortezomib therapy for extramedullary an era of novel induction drugs. Bone Marrow Transpl. 2019;
relapse of myeloma after autologous and non-myeloablative 54:1089–93.
allogeneic transplantation. Haematologica. 2005;90:278–9. 95. Katodritou E, Terpos E, Delimpasi S, Kotsopoulou M, Michalis
80. Ito C, Aisa Y, Mihara A, Nakazato T. Lenalidomide is effective E, Vadikolia C, et al. Real-world data on prognosis and outcome
for the treatment of bortezomib-resistant extramedullary disease of primary plasma cell leukemia in the era of novel agents: a
M. Bhutani et al.

multicenter national study by the Greek Myeloma Study Group. 104. Jurczyszyn A, Grzasko N, Gozzetti A, Czepiel J, Cerase A,
Blood Cancer J. 2018;8:31. Hungria V, et al. Central nervous system involvement by
96. Jurczyszyn A, Radocha J, Davila J, Fiala MA, Gozzetti A, multiple myeloma: a multi-institutional retrospective study of
Grzasko N, et al. Prognostic indicators in primary plasma cell 172 patients in daily clinical practice. Am J Hematol. 2016;
leukaemia: a multicentre retrospective study of 117 patients. Br J 91:575–80.
Haematol. 2018;180:831–9. 105. Gozzetti A, Cerase A, Lotti F, Rossi D, Palumbo A, Petrucci
97. Ganzel C, Rouvio O, Avivi I, Magen H, Jarchowsky O, Herzog MT, et al. Extramedullary intracranial localization of multiple
K, et al. Primary plasma cell leukemia in the era of novel agents myeloma and treatment with novel agents: a retrospective survey
for myeloma - a multicenter retrospective analysis of outcome. of 50 patients. Cancer. 2012;118:1574–84.
Leuk Res. 2018;68:9–14. 106. Chen CI, Masih-Khan E, Jiang H, Rabea A, Cserti-Gazdewich C,
98. Jung SH, Lee JJ, Kim K, Suh C, Yoon DH, Min CK, et al. The Jimenez-Zepeda VH, et al. Central nervous system involvement
role of frontline autologous stem cell transplantation for primary with multiple myeloma: long term survival can be achieved with
plasma cell leukemia: a retrospective multicenter study radiation, intrathecal chemotherapy, and immunomodulatory
(KMM160). Oncotarget. 2017;8:79517–26. agents. Br J Haematol. 2013;162:483–8.
99. Iriuchishima H, Ozaki S, Konishi J, Matsumoto M, Murayama 107. Lee D, Kalff A, Low M, Gangatharan S, Ho P, Bajel A, et al.
K, Nakamura F, et al. Primary plasma cell leukemia in the era of Central nervous system multiple myeloma-potential roles for
novel agents: a multicenter study of the japanese society of intrathecal therapy and measurement of cerebrospinal fluid light
myeloma. Acta Haematol. 2016;135:113–21. chains. Br J Haematol. 2013;162:371–5.
100. Drake MB, Iacobelli S, van Biezen A, Morris C, Apperley JF, 108. Abdallah AO, Atrash S, Shahid Z, Jameel M, Grazziutti M,
Niederwieser D, et al. Primary plasma cell leukemia and auto- Apewokin S, et al. Patterns of central nervous system involve-
logous stem cell transplantation. Haematologica. 2010;95:804–9. ment in relapsed and refractory multiple myeloma. Clin Lym-
101. Mahindra A, Kalaycio ME, Vela-Ojeda J, Vesole DH, Zhang MJ, phoma Myeloma Leuk. 2014;14:211–4.
Li P, et al. Hematopoietic cell transplantation for primary plasma 109. Paludo J, Painuly U, Kumar S, Gonsalves WI, Rajkumar V,
cell leukemia: results from the center for international blood and Buadi F, et al. Myelomatous Involvement of the Central Nervous
marrow transplant research. Leukemia. 2012;26:1091–7. System. Clin Lymphoma Myeloma Leuk. 2016;16:644–54.
102. Nooka AK, Kaufman JL, Muppidi S, Langston A, Heffner LT, 110. Mussetti A, Dalto S, Montefusco V. Effective treatment of
Gleason C, et al. Consolidation and maintenance therapy with pomalidomide in central nervous system myelomatosis. Leuk
lenalidomide, bortezomib and dexamethasone (RVD) in high- Lymphoma. 2013;54:864–6.
risk myeloma patients. Leukemia. 2014;28:690–3. 111. Vicari P, Ribas C, Sampaio M, Arantes AM, Yamamoto M,
103. Katodritou E, Terpos E, Kastritis E, Delimpasis S, Symeonidis Filho JB, et al. Can thalidomide be effective to treat plasma cell
AS, Repousis P, et al. Lack of survival improvement with novel leptomeningeal infiltration? Eur J Haematol. 2003;70:198–9.
anti-myeloma agents for patients with multiple myeloma and 112. Badros A, Singh Z, Dhakal B, Kwok Y, MacLaren A,
central nervous system involvement: the Greek Myeloma Study Richardson P, et al. Marizomib for central nervous system-
Group experience. Ann Hematol. 2015;94:2033–42. multiple myeloma. Br J Haematol. 2017;177:221–5.

You might also like