You are on page 1of 14

Leukemia & Lymphoma

ISSN: 1042-8194 (Print) 1029-2403 (Online) Journal homepage: http://www.tandfonline.com/loi/ilal20

Recent advances in the management of multiple


myeloma: clinical impact based on resource-
stratification. Consensus statement of the Asian
Myeloma Network at the 16th international
myeloma workshop

Daryl Tan, Jae Hoon Lee, Wenming Chen, Kazuyuki Shimizu, Jian Hou, Kenshi
Suzuki, Weerasak Nawarawong, Shang-Yi Huang, Chor Sang Chim, Kihyun
Kim, Lalit Kumar, Pankaj Malhotra, Wee Joo Chng, Brian Durie & for the
Asian Myeloma Network

To cite this article: Daryl Tan, Jae Hoon Lee, Wenming Chen, Kazuyuki Shimizu, Jian Hou,
Kenshi Suzuki, Weerasak Nawarawong, Shang-Yi Huang, Chor Sang Chim, Kihyun Kim, Lalit
Kumar, Pankaj Malhotra, Wee Joo Chng, Brian Durie & for the Asian Myeloma Network (2018):
Recent advances in the management of multiple myeloma: clinical impact based on resource-
stratification. Consensus statement of the Asian Myeloma Network at the 16th international
myeloma workshop, Leukemia & Lymphoma, DOI: 10.1080/10428194.2018.1427858

To link to this article: https://doi.org/10.1080/10428194.2018.1427858

View supplementary material Published online: 02 Feb 2018.

Submit your article to this journal View related articles

View Crossmark data

Full Terms & Conditions of access and use can be found at


http://www.tandfonline.com/action/journalInformation?journalCode=ilal20
LEUKEMIA & LYMPHOMA, 2018
https://doi.org/10.1080/10428194.2018.1427858

REVIEW

Recent advances in the management of multiple myeloma: clinical impact


based on resource-stratification. Consensus statement of the Asian Myeloma
Network at the 16th international myeloma workshop
Daryl Tana,b, Jae Hoon Leec, Wenming Chend, Kazuyuki Shimizue, Jian Houf, Kenshi Suzukig,
Weerasak Nawarawongh, Shang-Yi Huangi, Chor Sang Chimj, Kihyun Kimk, Lalit Kumarl, Pankaj Malhotram,
Wee Joo Chngn,o and Brian Duriep; for the Asian Myeloma Network
a
Raffles Cancer Center, Raffles Hospital, Singapore; bDepartment of Hematology, Singapore General Hospital, Singapore; cGil Hospital,
Gachon University, Incheon, South Korea; dBeijing Chaoyang Hospital, Capital Medical University, Beijing, China; eHigashi Nagoya
National Hospital, National Hospital Organization, Nagoya, Japan; fDepartment of Haematology, Changzheng Hospital, The Second
Military Medical University, Shanghai, China; gDepartment of Hematology, Japanese Red Cross Medical Center, Tokyo, Japan; hChiang
Mai Medical School, Maung, Thailand; iNational Taiwan University Hospital, Taipei, Taiwan; jQueen Mary Hospital, University of Hong
Kong, Hong Kong, China; kSamsung Medical Center, Sungkyunkwan University, Seoul, South Korea; lDepartment of Medical Oncology,
Institute Rotary Cancer Hospital, All India Institute of Medical Sciences, New Delhi, India; mDepartment of Internal Medicine,
Postgraduate Institute of Medical Education and Research, Chandigarh, India; nCancer Science Institute of Singapore, National
University of Singapore, Singapore; oDepartment of Haematology-Oncology, National University Cancer Institute of Singapore
National University Health System, Singapore; pCedars-Sinai Comprehensive Cancer Center, Los Angeles, CA, USA

ABSTRACT ARTICLE HISTORY


Predicated on our improved understanding of the disease biology, we have seen remarkable Received 20 September 2017
advances in the management of multiple myeloma over the past few years. Recently approved Revised 25 December 2017
drugs have radically transformed the treatment paradigm and improved survivals of myeloma Accepted 9 January 2018
patients. The progress has necessitated revision of the diagnostic criteria, risk-stratification and
KEYWORDS
response definition. The huge disparities in economy, healthcare infrastructure and access to Myeloma; resource-
novel drugs among different Asian countries will hinder the delivery of optimum myeloma care stratified; novel agents
to patients managed in resource-constrained environments. In the light of the tremendous
recent changes and evolution in myeloma management, it is timely that the resource-stratified
guidelines from the Asian Myeloma Network be revised to provide updated recommendations
for Asia physicians practicing under various healthcare reimbursement systems. This review will
highlight the most recent advances and our recommendations on how they could be integrated
in both resource-abundant and resource-constrained facilities.

Background and introduction novel drugs has resulted in remarkable progress in mye-
loma [5]. Since the publication of our guidelines, six
Multiple myeloma is the second most frequent hema- new drugs that are radically transforming the treatment
tological malignancy, accounting for 1% of all new paradigm have been approved by the Food and Drug
cancer incidences and mortality globally [1]. The Asian Administration of America (FDA). Recent advances have
Myeloma Network (AMN) has reported rising incidence necessitated revision of disease definition, risk-stratifica-
of myeloma in Asia and that no major difference in tion and prognostication [6–8]. More stringent and pre-
clinical or biological characteristics exists between cise criteria for response definition that include minimal
Asian and Western cases [2]. In view of the huge dis- residual disease (MRD) measurement and treatment
parity in healthcare across Asia, and with an aim to algorithms that integrate high-dose therapy with
provide a practical framework for planning resource- autologous stem cell transplantation (HDT-ASCT), tar-
appropriate myeloma care, the AMN formulated a set geted therapies and immunotherapy have also evolved
of resource-stratified recommendations based on the [9]. Despite phenomenal breakthroughs, myeloma
framework established by the Breast Health Global remains incurable. Longer survivals would now translate
Initiative where health-care resources were stratified to higher cumulative prevalence, taxing prevailing
according to a four-tiered system on basis of available healthcare systems further. Clinical outcomes of
resources (Table 1) [3,4]. Of late, the introduction of patients treated under resource-abundant and

CONTACT Daryl Tan tan_daryl@rafflesmedical.com Raffles Cancer Center, 585 North Bridge Road, #13-00 Raffles Hospital, 88770 Singapore,
Singapore
ß 2018 Informa UK Limited, trading as Taylor & Francis Group
2 D. TAN ET AL.

Table 1. Four-tiered system based on available resources rele- to one defined by biomarkers where serum free light
vant to program implementation [3]. chains (SFLC) assay and accurate quantification of
Basic level Core resources or fundamental services that are abso- bone marrow plasma cells (BMPC) now play vital roles
lutely necessary for any cancer system to function;
basic-level services typically are applied in a single especially for patients with high-risk smoldering mye-
clinical interaction loma [6]. These changes are based on the observations
Limited level Second-tier resources or services that are intended to
produce major improvements in outcome such as that having BMPC of 60%, involved to uninvolved
improved survival, and are attainable with limited SFLC ratio of 100 or the presence of more than one
financial means and modest infrastructure; limited-
level services may involve single or multiple clinical focal lesion with whole-body MRI was associated with
interactions a 70–90% risk of progression to myeloma [11–13]. In
Enhanced level Third-tier resources or services that are optional in a
resource-constrained setting but are important and addition, early intervention in high-risk asymptomatic
should produce further improvements in outcome patients has been shown to extend survival [14,15].
and increase the number and quality of therapeutic
options and choices for patients The preemptive approach of treating these patients
Maximal level High-level resources or services that may be used in with ultra-high-risk smoldering myeloma could also
some high-resource countries and/or may be rec-
ommended by myeloma care guidelines that do prevent the onset of organ damage which may further
not adapt to resource constraints but that nonethe- escalate healthcare costs. To avoid missing the diagno-
less should be considered a lower priority than
those resources or services listed in the basic, lim-
sis of myeloma among asymptomatic patients, accur-
ited, or enhanced categories on the basis of ate enumeration of BMPC is important. In clinical
extreme cost and/or impracticality for broad use in
a resource-limited environment; to be useful, max-
practice, BMPC estimation is typically higher based on
imal-level resources typically depend on the exist- trephine biopsy with CD138 immunohistochemical
ence and functionality of all lower level resources staining compared to estimation on aspirate smear or
enumeration by flow cytometry [16]. As simple H&E
resource-constrained facilities will continue to be more staining may frequently underestimate BMPC count,
disparate. In the light of rapid recent changes, the AMN immunohistochemical staining should not be omitted
under the auspice of the International Myeloma for cost containment. Ambiguous cases could be
Foundation convened a meeting during the 16th referred for opinion at more equipped facilities.
International Myeloma Workshop to discuss the clinical The definitions of the ‘CRAB’ criteria are recently
impact of recent advances and provided consensus rec- further fine-tuned. The IMWG recommends that meas-
ommendations based on resource-stratification. ured or estimated glomerular filtration rates <40 mL/
min be used instead of a fixed serum creatinine con-
Resource-stratified consensus framework centration (>173 mmol/L or >2 mg/dL) as the latter
may be affected by weight, age and gender [6]. For
As most countries in Asia are in the low- or middle- unclear cases where other concomitant comorbidities
income category, the latest International Myeloma
may contribute to renal impairment, renal biopsy is
Working Group (IMWG) recommendations may not be
recommended [17]. Newer imaging techniques have
implementable. A resource-constrained center must
now shown greater sensitivity than radiographic bone
invest in facilities for myeloma care in a cost-effective,
survey for defining myeloma bone disease (1 sites of
stepwise fashion to achieve the best results with the
osteolytic bone destruction 5 mm in size) [13,18]. The
most rational allocation of resources. As myeloma is
revised IMWG criteria now recognize positron-emission
rare, patients treated at higher-volume facilities report-
tomography-computed tomography (PET-CT) or low-
edly have a lower risk of mortality compared with
dose whole-body CT as valid methods to assess for
those treated at lower-volume facilities [10]. Basic
lytic bone lesions. It should be stressed that increased
healthcare facilities will lack volume, expertise and
uptake on PET-CT alone is not adequate for the diag-
access to novel drugs. In order to avoid preventable
nosis of myeloma and the CT portion of the PET-CT
early mortalities, especially among patients with high-
scan is more important for assessment of osteolytic
risk disease, myeloma patients should not be managed
bone destruction. Presence of osteoporosis or verte-
in settings with only basic facilities. Hence, our current
bral compression fractures alone in the absence of
recommendations will be based on a three-tiered sys-
tem (limited/enhanced/maximum). lytic lesions is no longer accepted as evidence of bone
disease. Whole-body or axial MRI may be essential
especially if the skeletal survey or PET-CT doesn’t show
Diagnosis of multiple myeloma any bone lesion, or when an assessment for extraoss-
In 2014, the IMWG revised the definition of myeloma eous soft tissue masses is needed [13]. Physicians will
from being a disease defined primarily by symptoms need to appreciate the limitations of the various
RESOURCE-STRATIFIED MANAGEMENT OF MYELOMA 3

Table 2. Resource-adapted risk stratification.


Low risk Intermediate risk High risk
Limited ISS I without non-hyperdiploid karyo- Neither low nor high risk ISS III plus non-hyperdiploid karyotype or
type and with normal LDH high LDH
Enhanced R-ISS I and age < 55 R-ISS II R-ISS III
Maximum R-ISS I and age < 55 with standard- Neither low or high risk  R-ISS III
risk signature on GEP or NGS  High-risk signature with GEP or NGS
GEP: gene expression profiling; NGS: next generation sequencing; ISS: International Staging System; R-ISS: revised-ISS.
iFISH facility may not be available.
GEP or NGS complements, but does not substitute the R-ISS in risk-stratification.

imaging modalities and know when to refer patients if a non-hyperdiploid karyotype is detected [19,27].
for more complex imaging. The critical iFISH panel done at centers with enhanced
facilities should include probes for 17p13 deletion,
t(4;14) and t(14;16) as they are part of the R-ISS.
Risk stratification
However, an extended panel incorporating t(11;14),
Multiple myeloma is clinically heterogeneous with t(14;20), gain(1q), del(1p) and del(13q) should be con-
median survivals ranging from 3 to more than 10 sidered whenever possible, especially if the patient is
years. Risk-stratification is hence vital for prognostica- accrued in clinical trial as it is important to evaluate
tion, defining treatment strategies and comparison of for cytogenetic subgroups that may have preferential
outcomes between therapeutic trials. As novel treat- sensitivity to certain novel agents [28,29]. A subopti-
ments may alter prognosis, prognostication systems mal response to induction and relapse within a year of
will have to evolve with changes in treatment para- HDT-ASCT are also accurate predictors of dismal out-
digms. The AMN previously recommended whenever comes [30,31].
possible, the use of an integrated model of
International Staging System (ISS) with interphase
Disease monitoring and response criteria
fluorescent in situ hybridization (iFISH) [4,19–21]. This
was recently combined with serum lactate dehydro- We previously recommended that response to treat-
genase (LDH) in the Revised-ISS (R-ISS) [7,22]. An ele- ment be defined according to IMWG uniform response
vated LDH level is indicative of more aggressive criteria and evaluation of MRD be done only in context
disease with higher proliferation rate and/or the pres- of research [3,32]. While this recommendation remains
ence of extramedullary involvement [23]. With an valid especially in resource-constrained facilities where
improved prognostic power that has been validated, access to novel agents and HDT-ASCT is still not univer-
the R-ISS is currently the preferred prognostication sal and a conventional CR remains a desirable treatment
tool [7]. Several gene expression signatures which goal that is attained by the minority, it may be inad-
have been associated with high-risk disease are now equate in enhanced/maximal systems where response
available as commercial platforms [24–26]. Specific assessment needs to evolve with the treatment advan-
combinations of high-risk signatures with the ISS and ces. Novel drugs have tremendously improved the rates
iFISH were demonstrated to further enhance prognos- and depth of response such that higher rates of CR are
tic power [25,26]. Large scale sequencing studies have now expected. New response grades that can further
become feasible with the advent of next generation discriminate responses that are deeper are needed.
sequencing (NGS) and mutations detected by NGS Based on multiparameter flow cytometry (MFC) and
have been shown to further compliment current prog- gene sequencing, the IMWG has defined new response
nostication methods [26]. Although highly precise, categories of MRD-negativity, with or without imaging-
genomic approaches may have limited usage in rou- based absence of extramedullary disease, to allow uni-
tine practice because of their complexity and costs. form reporting within and outside of clinical trials (Table
In facilities with limited resources, the ISS and the 3) [9]. It is now apparent that benefits of CR come
LDH level should be minimal risk-stratification parame- mainly from the MRD-negative cases [33]. A recent
ters since they are based on universally obtainable meta-analysis reported that newly diagnosed patients
laboratory measures (Table 2). Although the utilization who were MRD-negative after upfront therapy had sig-
of conventional metaphase karyotyping has been nificantly better PFS and OS than counterparts who
declining due to its lack of sensitivity compared to were MRD-positive despite being in CR [34]. Currently,
iFISH, it remains the most easily performed cytogenetic MRD assessment is being implemented in most clinical
test with resource limitation. In the present era, it still trials and many academic centers are applying it rou-
provides important prognostic information, especially tinely. Although its role in informing treatment decisions
4 D. TAN ET AL.

Table 3. IMWG MRD criteria (requires a complete response). criteria for PET-CT reporting for myeloma with the
Sustained MRD-negative MRD negativity in the marrow (NGF or incorporation of semi-quantitative SUV evaluations
NGS, or both) and by imaging as
defined below, confirmed minimum of
[41]. In the near future, newer imaging technologies
one year apart. Subsequent evaluations such as PET-MRI which is undergoing clinical evalua-
can be used to further specify the dur- tions may provide more precise information without
ation of negativity (e.g. MRD-negative
at five years) radiation exposure [42,43].
Flow MRD-negative Absence of phenotypically aberrant clonal Therapeutic monoclonal antibodies like daratumu-
plasma cells by NGF‡ on bone marrow
aspirates using the EuroFlow standard mab may interfere with monoclonal protein assessment
operation procedure for MRD detection and affect the accuracy of CR assignment [44]. Mass
in multiple myeloma (or validated
equivalent method) with a minimum spectrometry-based techniques are able to discriminate
sensitivity of 1 in 105 nucleated cells the different proteins based on their masses and pre-
or higher
Sequencing DNA sequencing of bone marrow aspi-
vent the inaccuracies, but are not available in most cen-
MRD-negative rates using the LymphoSIGHT platform ters in Asia [45]. Anti-idiotype antibodies that neutralize
(or validated equivalent method) with the offending drug (e.g. DARA IFE reflex assay) will be
a minimum sensitivity of 1 in 105
nucleated cells or higher commercially available and should be considered for
Imaging plus MRD negativity as defined by NGF or NGS patients receiving monoclonal antibody therapy [46]. In
MRD-negative plus disappearance of every area of
increased tracer uptake found at base- addition, MRD testing by MFC may also be affected
line or a preceding PET/CT or decrease since CD38 is a critical surface marker exploited in MFC.
to less than mediastinal blood pool
SUV or decrease to less than that of Specific CD38 antibody clones or reagents, together
surrounding normal tissue with the most sensitive CD138-fluorochrome conju-
MRD: minimal residual disease; NGF: next generation flow; NGS: next gen- gates such as the current EuroFlow 2 tube eight-color
eration sequencing.
panel MFC platform may mitigate the problem and
allow treatment-independent assessment of MRD [9].
is yet to be defined, it may be vital for assessment of
treatment efficacy and prognostication [33].
Additionally, MRD may eventually be used as a regula- Front-line treatment: patients eligible for
tory endpoint for drug approvals [35]. transplantation
The two main methods for assessing MRD, MFC and For newly diagnosed and physically fit patients
molecular sequencing, differ in various aspects, which younger than 65–70 years, HDT-ASCT after induction
affect their clinical applicability. Next-generation multi- therapy remains standard [47]. The incorporation of
color flow cytometry (NGF) which utilizes at least eight novel agents into induction regimens further improves
markers has a sensitivity of 105 to 106 and up to the survival outcomes [48,49]. Table 4 summarizes the
30% of patients found to be MRD-negative with con- recommended front-line treatment options based on
ventional MFC may become MRD-positive with NGF resource availability. A three-drug combination that
[33,36]. Assessing MRD with molecular methods like includes a second generation IMiD like lenalidomide
allele-specific oligonucleotide real-time quantitative and a proteasome inhibitor could be optimal, but if
PCR (ASO RQ-PCR) has high sensitivity of up to 105, reimbursement disallows, any bortezomib-based triplet
but may be technically complex and is labor-intensive will be reasonable as the addition of a third agent like
[37]. More recently, NGS based on high-throughput thalidomide, cyclophosphamide or doxorubicin to bor-
sequencing for clonally rearranged immunoglobulin tezomib-dexamethasone has consistently shown
heavy chain and/or kappa light chain sequences may higher response rates [50]. The Myeloma XI study has
find wider applicability [38,39]. With MFC, samples will recently demonstrated that for patients who failed to
have to be processed within 24–48 h, but results may achieve better than a PR with initial therapy of either
be available immediately. With NGS however, samples cyclophosphamide/lenalidomide/dexamethasone (CRD)
need not be processed immediately and hence or cyclophosphamide/thalidomide/dexamethasone
patients not treated in academic centers may still get (CTD), employing additional induction with bortezo-
MRD assessment via commercial platforms. Monitoring mib/cyclophosphamide/dexamethasone (VCD) prior to
of MRD also includes an assessment of the extrame- HDT-ASCT deepened response prior to and after HDT-
dullary compartment by PET-CT especially since extra- ASCT and improved PFS [51]. This approach could be
medullary myeloma is now increasingly encountered considered in centers where upfront use of novel
with extended survival of patients. A negative PET-CT agents is not standard for all patients.
after HDT-ASCT is associated with superior survival Recent studies comparing upfront HDT-ASCT versus
[40]. There are ongoing attempts to standardize protracted course of novel agents with a deferred
Table 4. Resource-adapted recommendations by the AMN.
Resource level Limited Enhanced Maximal
Initial assessment  History and physical examination  Low-dose whole-body CT or
 Complete blood counts and differentials  PET-CT
 Peripheral blood smear for Rouleaux formation and circulating plasma cells  Whole-body MRI for ambiguous cases
 Chemistry screen including calcium, blood urea nitrogen, creatinine and albumin
 24 hour urine total protein and creatinine clearance
 Radiographic skeletal survey
 Electrocardiogram
 CT or MRI scan if indicated for assessment of extramedullary disease
 MUGA or echocardiography scan if anthracycline to be used
Tests to establish diagnosis  Serum and urine protein electrophoresis
 SFLC assay
 BMA and trephine biopsy
 Immunofixation
 Immunophenotyping by flow cytometry or immunohistochemistry
Staging and Risk stratification  ISS Interphase FISH  GEP
 LDH Minimum:  NGS
 Metaphase karyotyping -t(4;14) Complements
 Early Relapse/Progression (within 1 year of transplant) - t(14;16) R-ISS, but not mandatory
-17p13 del
Extended panel:
-t(11;14)
- t(14;20)
- gain(1q21)
- del(1p)
- del(13q
Response measurement and  Serum and urine protein electrophoresis  4- or 6-color MPF þ  NGF þ
disease monitoring  BMA and trephine biopsy  PET-CT þ  NGS þ
þ
 SFLC assay MRD assessment for deeper response  ASO RQ-PCR þ
þ
 MRI or CT scans for plasmacytoma or extramedullary disease evaluation and prognostication, not MRD assessment for deeper response
to inform treatment decisions evaluation and prognostication, not
to influence treatment decisions
Treatment for transplant-eligible  Cyclophosphamide/THAL/DEX  BTZ/LEN/DEX  Clinical trials
-Consider BTZ/Cyclophosphamide/DEX before HDT/ASCT if  PR  BTZ/Cyclophosphamide/DEX
 THAL/DEX  BTZ/Adriamycin/DEX
 DVd  BTZ/THAL/DEX
Mobilization:  BTZ/DEX
 G-CSF with or without prior cyclophosphamide chemotherapy  Rd
Mobilization:
 Plerixafor can be considered in
mobilization failures
Treatment for transplant-ineligible  MPT  Continuous Rd  Clinical trials
 THAL/DEX  BTZ/LEN/DEX
 Cyclophosphamide/THAL/Dex  VMP
 DVd  VMPT-VT
 BTZ/Dex
 BTZ/Cyclophosphamide/DEX
 BTZ/THAL/DEX
 BTZ/Adriamycin/DEX
RESOURCE-STRATIFIED MANAGEMENT OF MYELOMA

Treatment for relapsed/  THAL/DEX  CFZ/DEX  Clinical trials


refractory myeloma  DT-PACE  CFZ/LEN/DEX
(continued)
5
6 D. TAN ET AL.

HDT-ASCT at first relapse have informed that the for-

MPT: melphalan/prednisolone/thalidomide; DVd: dexamethasone/vincristine/liposomal doxorubicin; DEX: dexamethasone; LEN: lenalidomide; BTZ: bortezomib; Pano: panobinostat; VMP: bortezomib/melphalan/pred-
nisolone; VMPT-VT: bortezomib/melphalan/prednisolone/thalidomide and bortezomib/thalidomide maintenance; VP-V: bortezomib/prednisolone and bortezomib maintenance; Rd: lenalidomide with lower-dose dexa-
mer approach results in superior CR, MRD-negativity
rates and PFS [52–54]. Although the frontline induction
regimen differed in the studies (lenalidomide/dexa-
Maximal

methasone [Rd], VCD and lenalidomide/bortezomib/


dexamethasone [VRD], respectively), they represent the
most commonly used novel induction combinations
currently. Despite the absence of an OS difference, it is
clear that novel agents are still not able to replace the
role of HDT-ASCT. Deferring HDT-ASCT would certainly
escalate healthcare costs and some patients may even-
tually become transplant-ineligible. Notwithstanding,
the IFM study has shown that regardless of HDT-ASCT,
POM/Cyclophosphamide/DEX

patients attaining MRD-negativity had similar superior


outcomes. As HDT-ASCT is still a very cost-effective
Enhanced

modality, whenever possible, patients treated in non-


transplant centers should always be referred for it.
DARA/LEN/DEX
DARA/BTZ/DEX
POM/CFZ/DEX
POM/BTZ/DEX

ELO/LEN/DEX
IXA/LEN/DEX

Currently, the role of tandem ASCT which was previ-


HDT/ASCT
POM/DEX

Pano/BTZ

ously found to be beneficial for patients who failed to


DARA

achieve at least a very good partial response (VGPR)


remains to be defined [55–58]. In a meta-analysis












based on phase III European studies, tandem trans-


plantation was shown to be beneficial for patients
with high-risk disease while the recently reported EMN
study suggests that tandem ASCT appears to be super-
ior in extending PFS compared with single ASCT after
Resources at the higher levels always depend on the existence and functionality of all lower-level resources.

induction therapy with limited cycles of VCD regimen


[56,57]. The phase III StaMINA study, however, indi-
cated that tandem ASCT followed by lenalidomide
maintenance gives similar outcomes as single ASCT
DEX/Cyclophosphamide/etoposide/cisplatin (DCEP)

followed by lenalidomide maintenance [58]. Most


methasone; CFZ: carfilzomib; POM: pomalidomide; DARA: daratumumab; ELO: elotuzumab.

patients in the study received VRD regimen as induc-


Limited

tion therapy for 2 to 12 months. Moreover, the tan-


dem-transplant cohort suffered from a large drop-out
rate, in part, reflecting actual patients’ reservation
against a second transplant. As such, tandem-ASCT
BTZ/Cyclophosphamide/DEX

should not be systemically applied as a routine.


Clarithromycin/LEN/DEX
Bendamustine/LEN/DEX
Bendamustine/BTZ/DEX

Nonetheless, for selected patients with high-risk dis-


BTZ/Adriamycin/DEX

ease or suboptimal response after the first ASCT, it


BTZ/THAL/DEX

Bendamustine
BTZ/LEN/DEX
BTZ/DT-PACE

may be an option especially in facilities where lenali-


BTZ/Doxil

domide and proteasome inhibitor cannot be given


together in induction or consolidation therapy.
RD

The StaMINA study also does not support the use















of consolidation after HDT/ASCT. Similarly, although


the role of consolidation remains to be defined, it
could be applied at the discretion of the treating phys-
ician after taking into consideration the cytogenetic
Table 4. Continued

risk, response attained after HDT-ASCT and resource


availability [58]. Based on major trials and meta-ana-
lysis, lenalidomide maintenance after a single ASCT
Resource level

clearly leads to better PFS and OS [59–61].


Lenalidomide lacks the neurotoxicity seen with pro-
longed use of thalidomide. However, the associated
RESOURCE-STRATIFIED MANAGEMENT OF MYELOMA 7

increased risk of secondary cancers after HDT-ASCT treatment subgroups. Those who are ‘very fit’ or ‘fit’
mandates a thorough discussion with patients before may receive many of the regimens described for trans-
its commencement. The costs of prolonged treatment plant-eligible patients where three-drug combinations
with lenalidomide can be prohibitive in most Asian are preferred while ‘unfit’ patients should receive a
countries. Although maintenance with thalidomide two-drug regimen with an aim to achieve the max-
may be more financially feasible, precautions must be imum response with minimal treatment-related toxicity
taken against the development of significant neurotox- [67,69]. The various bortezomib and lenalidomide com-
icity which could preclude the use of bortezomib dur- binations previously recommended will still remain
ing subsequent relapses [62]. Bortezomib maintenance valid [4]. The recent SWOG S0777 study however, has
may considered for patients with t(4;14) [29]. Due to demonstrated that VRD improved PFS and OS with an
the high treatment-related mortality and conflicting acceptable risk-benefit profile and could be the pre-
reports from prospective studies allogeneic stem-cell ferred three-drug combination if reimbursement per-
transplantation in frontline setting should be done mits [70].
only in the context of clinical trials [63]. For patients with resource constraints, the combin-
ation of melphalan/prednisolone/thalidomide (MPT)
Risk-adapted approach should be considered a minimal standard of care as it
has shown consistent improvements in PFS and even
Individualizing therapy based on cytogenetic risk OS [71]. The recently completed Frontline Investigation
remains a contentious issue and is largely dependent Of Lenalidomide/Dexamethasone Versus Standard
on the healthcare reimbursement system. Although Thalidomide (FIRST) trial showed that continuous Rd
optimal treatments for high-risk myeloma have yet to
given until disease progression was associated with a
be defined, prospective studies have informed that the
significant improvement in PFS and OS compared to
prognosis of high-risk patients may vary with the choice
fixed duration of Rd or MPT [72]. Adverse events and
of therapy [29]. In particular, the use of bortezomib-
subsequent hematologic malignancies were less fre-
based treatment for patients with t(4;14) has con-
quent with continuous Rd. Subgroup analysis of Asian
sistently shown improved outcomes while the risk
patients concluded that continuous Rd is safe and
associated with deletion 17p may be mitigated with
effective [73]. A separate randomized study has
regimens that contain pomalidomide, carfilzomib and
reported that alkylator-containing triplets of MPR and
daratumumab [48,64]. The hypothesis that high-risk
CPR were not superior to the alkylator-free doublet Rd,
patients are most at risk for clonal evolution if they lack
which was also associated with lower toxicities [74]. A
durable response to initial therapies now prompts the
network meta-analysis demonstrated that Rd was asso-
use of the most highly active combinations early to
ciated with significant PFS and OS advantage over
reduce the emergence of resistance [65]. Patients with
other first-line treatments including VMP, MPT and MP.
high-risk disease in enhanced or maximal systems
should hence actively enroll in frontline trials that Although the optimal duration for induction therapy
incorporate various next-generation novel agents or after achievement of maximal response is still
monoclonal antibodies where measurement of MRD is as unknown, data from the FIRST and other recent stud-
an endpoint. In resource-constrained facilities where ies have consistently shown that continuous therapy
frontline access to novel agents may not be uniform, risk- provide greater benefit including improved PFS2 (time
stratification may be a mean to triage resources and avail from random assignment until the second progression
novel agents to patients most at risk of early mortality [66]. or death) than fixed-duration therapy [72,75–77]. A
meta-analysis has raised concern that the risk of sec-
ondary primary malignancy may be higher in patients
Front-line treatment: patients ineligible for receiving lenalidomide with melphalan [78]. Taken
transplantation together, the data represent a paradigm shift away
Transplant ineligible patients form a heterogeneous from alkylator-based regimens given for a fixed dur-
group. Chronological age alone is not a reliable deter- ation to an alkylator-free regimen given continuously.
minant of functional reserve or the risk of complica- Thus, in enhanced or maximal systems where upfront
tions from therapy. A comprehensive geriatric access to bortezomib and lenalidomide may be univer-
assessment like the IMWG score should be applied sal, melphalan could be omitted whereas in facilities
routinely before therapy to define the frailty profile of with limited access to the two drugs, melphalan will
the patient [67,68]. Based on age, comorbidities and still remain an important backbone. Although using
frailty, elderly patients can be divided into three thalidomide or bortezomib as continuous therapy may
8 D. TAN ET AL.

still appear efficacious, this has to be balanced against pomalidomide/dexamethasone (Pd), bortezomib/dexa-
the risk of neurotoxicity [75,76,79,80]. methasone (Vd), or carfilzomib/dexamethasone (Kd)
are reasonable for frail patients who may not tolerate
triplet therapy. Salvage HDT-ASCT can be considered
Management of relapse and refractory
for transplant-eligible patients who have yet to receive
multiple myeloma
one or had a durable post-HDT-ASCT remission of
Indications for starting treatment were recently more than 18 months. When high-risk genetics are
defined by the IMWG [81]. Synergistic combinations absent, this may be a more economical option in first
that can overcome high-risk features and the continu- relapse compared to protracted courses of novel
ation of therapy till disease progression or the devel- agents [88].
opment of intolerable toxicity in order to suppress
clonal evolution are now widely accepted as the sal-
Second and subsequent relapses
vage strategies. This would inevitably compound treat-
ment costs and may not be feasible with resource Management now has to be balanced against the
limitation. If reimbursement precludes continuous ther- accumulation of toxicities from therapies. For patients
apy, the physician should strive to at least achieve a relapsing while receiving a doublet, it is prudent that
plateau before stopping therapy. The ability of certain a triplet salvage be used to overcome genomic
new agents to mitigate adverse cytogenetic risks instability [84,89,90]. Patients who have not been
would prompt the assessment for acquisition of new exposed to monoclonal antibodies should consider
mutations such as 17p deletion, 1q amplification or 1p one of the combinations like daratumumab/bortezo-
deletion [82]. Ability to manage the cumulative and mib/dexamethasone, daratumumab/lenalidomide/
emergent toxicities from therapies is also important to dexamethasone or elotuzumab/lenalidomide/dexa-
prevent premature discontinuation which will nega- methasone [91–93]. Combination of a bortezomib with
tively impact the eventual outcome [83]. Increased panobinostat may also be considered, but physicians
options now allow for a more systematic approach to need to be aware of the peculiar toxicities of the com-
relapsed/refractory disease. bination [94]. When rapid disease control is needed or
when patient develops plasma cell leukemia or extra-
medullary disease, intensive regimes like VDT-PACE
First relapse
(bortezomib/dexamethasone/thalidomide/cisplatin/
Patients with enhanced resources are more likely to be doxorubicin/cyclophosphamide/etoposide) can be
receiving maintenance or continuous therapy at considered [95]. As a result of clonal tides, it may be
relapse while those with limited resources are more reasonable to repeat the same agents previously used
likely to relapse in the absence of therapy. For the lat- [96]. Patients who have quadruple-refractory disease
ter group, the primary induction regimen may be (resistance to lenalidomide/pomalidomide/bortezo-
repeated if it accorded a durable remission of at least mib/carfilzomib) should receive salvage therapy that
six to nine months, whereas an alternative regimen incorporates a monoclonal antibody or enroll in a clin-
that incorporates at least one noncross resistant drug ical trial that explores other novel modalities. The vari-
should be considered if the remission duration with ous resource-appropriate options in the salvage
the prior regimen is short. Bortezomib or lenalidomide setting based on rationally designed clinical trials are
should be used if patients have never been exposed listed in Table 4.
to them. Patients with enhanced resources would have
been exposed to either or both of these two drugs
Drug access and clinical trials in Asia
and hence a triplet with at least one new class of
agent or a change to a next-generation agent from It is clear from recent studies that novel agents are
the same class should be considered [84,85]. Triplets, able to induce deep remissions and even MRD-nega-
whenever possible would be preferred to doublets as tivity, and gaining early access to them during relapse
they are associated with deeper remissions and better is key to prolonging survivals. However, drug access
survival outcomes. For patients treated under resource and healthcare reimbursement in many countries of
constraints where novel triplets may not be feasible, Asia are very limited [97]. For countries lacking
the addition of cyclophosphamide or clarithromycin is universal health coverage, only the affluent population
a consideration. Albeit with limited data, they may can afford novel drugs. Initiatives in the forms of
improve outcomes without substantially increasing named-patient program or patient-assistance program
costs and toxicities [86,87]. Doublets such as Rd, will be critical in availing drugs to patients [98] In
RESOURCE-STRATIFIED MANAGEMENT OF MYELOMA 9

some countries, availability of generics or bio-similar and in patients given rituximab. As such, entacavir
may help to defray costs of treatment, thereby allow- might be the preferred anti-viral prophylaxis [105,106].
ing more patients to be optimally treated [99].
Published experience with some of these drugs have
Conclusions
validated similar efficacy without increased toxicity
[100–102]. An important AMN initiative is the estab- The management of myeloma is continuously evolv-
lishment of a clinical trial network across Asia with the ing. Evidently, the key to improved survival outcomes
aims to avail novel drugs that may not yet be com- is the access to the latest quality diagnostics and novel
mercially available in Asian countries, validate drug therapeutics. Healthcare resources have to be opti-
responses and study peculiar patterns of adverse mized to achieve this goal. The role of clinicians and
events among Asian patients. Cross-border participa- researchers involved in global myeloma care should
tion is encouraged as this allows patients from coun- be one of leveling the playing field such that one’s
tries that lag behind in drug approvals or clinical birthplace does not determine one’s chance of surviv-
research to gain access to novel drugs. Physicians are ing a treatable illness like myeloma. The AMN is com-
referred to the AMN website, https://www.myeloma. mitted to bridging the gap between patients treated
org/research/asian-myeloma-network for the latest
under resource-abundant and resource-constrained
updates on drug approvals and AMN clinical trials in Asia.
facilities. We hope this resource-stratified framework,
used in conjunction with the available evidence, will
Supportive care help to improve healthcare services in Asia to better
manage myeloma.
Supportive care plays an even more important role
now as patients are living longer with the burden of
disease complications as well as the cumulative side Search strategy and selection criteria
effects of treatment. Although our previous consensus
Articles for this review were found through searches of
for adjunctive management of venothromboembolism,
PubMed, Medline, and references from relevant articles
skeletal complications, infections and treatment-emer-
gent peripheral neuropathy would still remain valid, by use of the search terms: ‘multiple myeloma,’ ’Asia,’
emerging concerns pertaining to specific new drugs ‘risk stratification,’ ’treatment’ and ‘cytogenetics.’ Trials
will need to be addressed. In particular, as hepatitis B with the highest quality of study design were selected.
virus (HBV) infection is highly endemic in Asia, a major Only studies of adults, written in English, and pub-
concern is the risk of its reactivation after the use of lished between January 1985 and April 2017 were
anti-CD38 therapy. The experience with rituximab in included.
lymphoma treatment has informed that a high risk of
HBV reactivation may be encountered even among Acknowledgments
patients who are HBsAg-negative, but anti-HBV core
antibody-positive [103,104]. This relates to a comprom- We thank Susie Novis, president of International Myeloma
Foundation (IMF), Dan Navid, vice president, global affairs of
ise of the B-cell associated humoral immunity when
IMF and Lisa Paik, senior vice president, clinical education
CD20 is targeted. Although there has so far been no and research initiatives of IMF, Xuan Lam, medical affairs
reported case of HBV reactivation, in part due to the assistant of IMF and Amirah Limayo, research project coord-
lack of exposure of these drugs in Asia, the theoretical inator, IMF for their administrative efforts making this AMN
risk of reactivation could be at least similar to that project possible.
seen with rituximab after plasma cells are targeted by
anti-CD38 therapy. In addition, the suppressive effect Potential conflict of interest: Disclosure forms provided
of anti-CD38 antibodies on regulatory T-cells may by the authors are available with the full text of this article
result in an exaggerated T-cell-mediated immuno- online at https://doi.org/10.1080/10428194.2018.1427858.
logical response and hepatic attack upon recovery
from immunesuppression. Hence, it is mandatory to References
screen for both HBsAg and anti-HBV core antibody for
[1] Ferlay J, Shin HR, Bray F, et al. Estimates of world-
all Asian patients who will be prescribed an anti-CD38
wide burden of cancer in 2008: GLOBOCAN 2008. Int
antibody. Pre-emptive prescription of prophylaxis and
J Cancer. 2010;127:2893–2917.
monitoring of HBV load should be accessible at all lev- [2] Kim K, Lee JH, Kim JS, et al. Clinical profiles of mul-
els of resources. Resistance to lamivudine has been tiple myeloma in Asia – An Asian Myeloma Network
reported especially in patients with high HBV titers study. Am J Hematol. 2014;89:751–756.
10 D. TAN ET AL.

[3] Anderson BO, Yip CH, Smith RA, et al. Guideline myeloma-related renal impairment. J Clin Oncol.
implementation for breast healthcare in low-income 2016;34:1544–1557.
and middle-income countries: overview of the Breast [18] Regelink JC, Minnema MC, Terpos E, et al.
Health Global Initiative Global Summit 2007. Cancer Comparison of modern and conventional imaging
2008;113:2221–2243. techniques in establishing multiple myeloma-related
[4] Tan D, Chng WJ, Chou T, et al. Management of mul- bone disease: a systematic review. Br J Haematol.
tiple myeloma in Asia: resource-stratified guidelines. 2013;162:50–61.
Lancet Oncol. 2013;14:e571–e581. [19] Fonseca R, Barlogie B, Bataille R, et al. Genetics and
[5] Kumar SK, Dispenzieri A, Lacy MQ, et al. Continued cytogenetics of multiple myeloma: a workshop
improvement in survival in multiple myeloma: report. Cancer Res. 2004;64:1546–1558.
changes in early mortality and outcomes in older [20] Greipp PR, San Miguel J, Durie BG, et al.
patients. Leukemia 2014;28:1122–1128. International staging system for multiple myeloma. J
[6] Rajkumar SV, Dimopoulos MA, Palumbo A, et al. Clin Oncol. 2005;23:3412–3420.
International Myeloma Working Group updated crite- [21] Avet-Loiseau H, Durie BG, Cavo M, et al. Combining
ria for the diagnosis of multiple myeloma. Lancet fluorescent in situ hybridization data with ISS staging
Oncol. 2014;15:e538–e548. improves risk assessment in myeloma: an
[7] Palumbo A, Avet-Loiseau H, Oliva S, et al. Revised International Myeloma Working Group collaborative
international staging system for multiple myeloma: a project. Leukemia 2013;27:711–717.
report from International Myeloma Working Group. J [22] Moreau P, Cavo M, Sonneveld P, et al. Combination
Clin Oncol. 2015;33:2863–2869. of international scoring system 3, high lactate
[8] Chng WJ, Dispenzieri A, Chim CS, et al. IMWG con- dehydrogenase, and t(4;14) and/or del(17p) identifies
sensus on risk stratification in multiple myeloma. patients with multiple myeloma (MM) treated with
Leukemia 2014;28:269–277. front-line autologous stem-cell transplantation at
[9] Kumar S, Paiva B, Anderson KC, et al. International high risk of early MM progression-related death. J
Myeloma Working Group consensus criteria for
Clin Oncol. 2014;32:2173–2180.
response and minimal residual disease assessment in
[23] Terpos E, Katodritou E, Roussou M, et al. High serum
multiple myeloma. Lancet Oncol. 2016;17:e328–e346.
lactate dehydrogenase adds prognostic value to the
[10] Go RS, Bartley AC, Crowson CS, et al. Association
international myeloma staging system even in the
between treatment facility volume and mortality of
era of novel agents. Eur J Haematol. 2010;85:
patients with multiple myeloma. J Clin Oncol
114–119.
2017;35:598–604. 3rd.
[24] Chng WJ, Chung TH, Kumar S, et al. Gene signature
[11] Kastritis E, Terpos E, Moulopoulos L, et al. Extensive
combinations improve prognostic stratification of
bone marrow infiltration and abnormal free light
multiple myeloma patients. Leukemia 2016;30:
chain ratio identifies patients with asymptomatic
1071–1078.
myeloma at high risk for progression to symptomatic
[25] Kuiper R, Mark van Duin Martin H, van Vlietet, et al.
disease. Leukemia 2013;27:947–953.
[12] Larsen JT, Kumar SK, Dispenzieri A, et al. Serum free Prediction of high- and low-risk multiple myeloma
light chain ratio as a biomarker for high-risk smol- based on gene expression and the International
dering multiple myeloma. Leukemia 2013;27: Staging System. Blood 2015;126:1996–2004.
941–946. [26] Walker BA, Boyle EM, Wardell CP, et al. Mutational
[13] Hillengass J, Fechtner K, Weber MA, et al. Prognostic spectrum, copy number changes, and outcome:
significance of focal lesions in whole-body magnetic results of a sequencing study of patients with newly
resonance imaging in patients with asymptomatic diagnosed Myeloma. J Clin Oncol. 2015;33:
multiple myeloma. J Clin Oncol. 2010;28:1606–1610. 3911–3920.
[14] Mateos M-V, Hernandez M-T, Giraldo P, et al. [27] Tan D, Teoh G, Lau LC, et al. An abnormal nonhyper-
Lenalidomide plus dexamethasone for high-risk diploid karyotype is a significant adverse prognostic
smoldering multiple myeloma. N Engl J Med. factor for multiple myeloma in the bortezomib era.
2013;369:438–447. Am J Hematol. 2010;85:752–756.
[15] Sigurdardottir EE, Turesson I, Lund SH, et al. The role [28] Shaji Kumar MD, Ravi Vij MD, Jonathan L, et al.
of diagnosis and clinical follow-up of monoclonal Venetoclax monotherapy for relapsed/refractory mul-
gammopathy of undetermined significance on sur- tiple myeloma: safety and efficacy results from a
vival in multiple myeloma. JAMA Oncol. 2015;1: phase I study. Blood 2016;128:488.
168–174. [29] Sonneveld P, Avet-Loiseau H, Lonial S, et al.
[16] Rajkumar SV, Fonseca R, Dispenzieri A, et al. Treatment of multiple myeloma with high-risk cyto-
Methods for estimation of bone marrow plasma cell genetics: a consensus of the International Myeloma
involvement in myeloma: predictive value for Working Group. Blood 2016;127:2955–2962.
response and survival in patients undergoing autolo- [30] Martinez-Lopez J, Blade J, Mateos MV, et al. Long-
gous stem cell transplantation. Am J Hematol. term prognostic significance of response in multiple
2001;68:269–275. myeloma after stem cell transplantation. Blood
[17] Dimopoulos MA, Sonneveld P, Leung N, et al. 2011;118:529–534.
International myeloma working group recommenda- [31] Majithia N, Rajkumar SV, Lacy MQ, et al. Early relapse
tions for the diagnosis and management of following initial therapy for multiple myeloma
RESOURCE-STRATIFIED MANAGEMENT OF MYELOMA 11

predicts poor outcomes in the era of novel agents. immunofi xation (IFE) and clinical assessment of M-
Leukemia 2016;30:2208–2213. protein response in multiple myeloma. Cancer Res.
[32] Rajkumar SV, Harousseau JL, Durie B, et al. 2014;74:2563.
Consensus recommendations for the uniform report- [47] Cavo M, Rajkumar SV, Palumbo A, et al. International
ing of clinical trials: report of the International Myeloma Working Group consensus approach to the
Myeloma Workshop Consensus Panel 1. Blood treatment of multiple myeloma patients who are
2011;117:4691–4695. candidates for autologous stem cell transplantation.
[33] Paiva B, van Dongen JJ, Orfao A. New criteria for Blood 2011;117:6063–6073.
response assessment: role of minimal residual dis- [48] Cavo M, Tacchetti P, Patriarca F, et al. Bortezomib
ease in multiple myeloma. Blood 2015;125: with thalidomide plus dexamethasone compared
3059–3068. with thalidomide plus dexamethasone as induction
[34] Landgren O, Iskander K. Modern multiple myeloma therapy before, and consolidation therapy after, dou-
therapy: deep, sustained treatment response and ble autologous stem-cell transplantation in newly
good clinical outcomes. J Intern Med. 2017;281: diagnosed multiple myeloma: a randomised phase 3
365–382. study. Lancet. 2010;376:2075–2085.
[35] Landgren O, Devlin S, Boulad M, et al. Role of MRD [49] Rosinol L, Oriol A, Teruel AI, et al. Superiority of bor-
status in relation to clinical outcomes in newly diag- tezomib, thalidomide, and dexamethasone (VTD) as
nosed multiple myeloma patients: a meta-analysis. induction pretransplantation therapy in multiple
Bone Marrow Transplant. 2016;51:1565–1568. myeloma: a randomized phase 3 PETHEMA/GEM
[36] Flores-Montero J, Saboja-Flores L, Paiva B, et al. Next study. Blood 2012;120:1589–1596.
generation flow for highly sensitive and standardized [50] Moreau P, Attal M, Facon T. Frontline therapy of
detection of minimal residual disease in multiple multiple myeloma. Blood 2015;125:3076–3084.
myeloma. Leukemia. 2017;31:2094–2103. [51] Jackson GH, Davies FE, Pawlyn C, et al. Response
[37] Bai Y, Wong KY, Fung TK, et al. High applicability of adapted induction treatment improves outcomes for
ASO-RQPCR for detection of minimal residual disease myeloma patients; results of the phase III Myeloma
in multiple myeloma by entirely patient-specific pri- XI study. Blood 2016;128:1854.
mers/probes. J Hematol Oncol. 2016;9:107. [52] Palumbo A, Cavallo F, Gay F, et al. Autologous trans-
[38] Avet-Loiseau H. Minimal residual disease by next- plantation and maintenance therapy in multiple
generation sequencing: pros and cons. Am Soc Clin myeloma. N Engl J Med. 2014;371:895–905.
Oncol Educ Book. 2015;35:e425–e430. [53] Michele C, Antonio P, Sonja Z, et al. Upfront autolo-
[39] Martinez-Lopez J, Lahuerta JJ, Pepin F, et al. gous stem cell transplantation (ASCT) versus novel
Prognostic value of deep sequencing method for agent-based therapy for multiple myeloma (MM): a
minimal residual disease detection in multiple mye- randomized phase 3 study of the European Myeloma
loma. Blood 2014;123:3073–3079. Network (EMN02/HO95 MM trial). J Clin Oncol.
[40] Zamagni E, Patriarca F, Nanni C, et al. Prognostic 2016;34:8000.
relevance of 18-F FDG PET/CT in newly diagnosed [54] Attal M, Lauwers-Cances V, Hulin C, et al.
multiple myeloma patients treated with up-front Lenalidomide, bortezomib, and dexamethasone with
autologous transplantation. Blood 2011;118: transplantation for myeloma. N Engl J Med.
5989–5995. 2017;376:1311–1320.
[41] Nanni C, Zamagni E, Versari A, et al. Image interpret- [55] Cavo M, Tosi P, Zamagni E, et al. Prospective,
ation criteria for FDG PET/CT in multiple myeloma: a randomized study of single compared with double
new proposal from an Italian expert panel. IMPeTUs autologous stem-cell transplantation for multiple
(Italian Myeloma criteria for PET USe). Eur J Nucl myeloma: bologna 96 clinical study. J Clin Oncol.
Med Mol Imaging. 2015;43:414–421. 2007;25:2434–2441.
[42] Fraioli F, Punwani S. Clinical and research applica- [56] Cavo M, Salwender H, Rosin ~ol L, et al. Double Vs sin-
tions of simultaneous positron emission tomography gle autologous stem cell transplantation after borte-
and MRI. Br J Radiol. 2014;87:20130464. zomib-based induction regimens for multiple
[43] Pawlyn C, Fowkes L, Otero S, et al. Whole-body diff myeloma: an integrated analysis of patient-level data
usion-weighted MRI: a new gold standard for assess- from phase European III studies. Blood 2013;122:767.
ing disease burden in patients with multiple mye- [57] Cavo M, Petrucci MT,D, Raimondo, et al. Upfront sin-
loma? Leukemia 2016;30:1446–1448. gle versus double autologous stem cell transplant-
[44] McCudden CR, Voorhees PM, Hainsworth SA, et al. ation for newly diagnosed multiple myeloma: an
Interference of monoclonal antibody therapies with intergroup, multicenter, phase iii study of the
serum protein electrophoresis tests. Clin Chem. European myeloma network (EMN02/HO95 MM Trial).
2010;56:1897–1899. Blood 2016;128:991.
[45] Walz S, Stickel JS, Kowalewski DJ, et al. The antigenic [58] Stadtmauer EA, Pasquini MC, Blackwell B, et al.
landscape of multiple myeloma: mass spectrometry Comparison of autologous hematopoietic cell trans-
(re)defines targets for T-cell-based immunotherapy. plant (autoHCT), bortezomib, lenalidomide (Len) and
Blood 2015;126:1203–1213. dexamethasone (RVD) consolidation with Len main-
[46] Axel AE, McCudden CR, Xie H, et al. Development of tenance (ACM), tandem Autohct with Len mainten-
clinical assay to mitigate daratumumab, an IgG1j ance (TAM) and Autohct with Len maintenance (AM)
monoclonal antibody, interference with serum for up-front treatment of patients with multiple
12 D. TAN ET AL.

myeloma (MM): primary results from the randomized [72] Benboubker L, Dimopoulos MA, Dispenzieri A, et al.
phase III trial of the blood and marrow transplant Lenalidomide and dexamethasone in transplant-ineli-
clinical trials network (BMT CTN 0702 – StaMINA gible patients with myeloma. N Engl J Med.
Trial). Blood 2016;128:LBA–LB1-LBA. 2014;371:906–917.
[59] Attal M, Lauwers-Cances V, Marit G, et al. [73] Lu J, Lee JH, Huang SY, et al. Continuous treatment
Lenalidomide maintenance after stem-cell transplant- with lenalidomide and low-dose dexamethasone in
ation for multiple myeloma. N Engl J Med. transplant-ineligible patients with newly diagnosed
2012;366:1782–1791. multiple myeloma in Asia: subanalysis of the FIRST
[60] McCarthy PL, Owzar K, Hofmeister CC, et al. trial. Br J Haematol. 2017;176:743–749.
Lenalidomide after stem-cell transplantation for mul- [74] Magarotto V, Bringhen S, Offidani M, et al. Triplet vs
tiple myeloma. N Engl J Med. 2012;366:1770–1781. doublet lenalidomide-containing regimens for the
[61] Attal M, Palumbo A, Holstein SA, et al. Lenalidomide treatment of elderly patients with newly diagnosed
(LEN) maintenance (MNTC) after high-dose melpha- multiple myeloma. Blood 2016;127:1102–1108.
lan and autologous stem cell transplant (ASCT) in [75] Palumbo A, Bringhen S, Larocca A, et al. Bortezomib-
multiple myeloma (MM): a meta-analysis (MA) of melphalanprednisone-thalidomide followed by main-
overall survival (OS). J Clin Oncol. 2016;34:8001. tenance with bortezomib thalidomide compared
[62] Ludwig H, Durie BGM, McCarthy P, et al. IMWG con- with bortezomib-melphalan-prednisone for initial
sensus on maintenance therapy in multiple mye- treatment of multiple myeloma: updated follow-up
loma. Blood 2012;119:3003–3015. and improved survival. J Clin Oncol. 2014;32:634–640.
[63] Lokhorst H, Einsele H, Vesole D, et al. International [76] Mateos MV, Oriol A, Martınez-Lo’pez J, et al.
Myeloma Working Group consensus statement Bortezomib, melphalan, and prednisone versus bor-
regarding the current status of allogeneic stem-cell tezomib, thalidomide, and prednisone as induction
transplantation for multiple myeloma. J Clin therapy followed by maintenance treatment with
Oncol.2010;28:4521–4530. bortezomib and thalidomide versus bortezomib and
[64] San Miguel JF, Schlag R, Khuageva NK, et al.
prednisone in elderly patients with untreated mul-
Bortezomib plus melphalan and prednisone for initial
tiple myeloma: a randomised trial. Lancet Oncol.
treatment of multiple myeloma. N Engl J Med. 2008;
2010;11:934–941.
359:906–917.
[77] Palumbo A, Gay F, Cavallo F, et al. Continuous ther-
[65] Dispenzieri A. Myeloma: management of the newly
apy versus fixed duration of therapy in patients with
diagnosed high-risk patient. Hematology Am Soc
newly diagnosed multiple myeloma. J Clin Oncol.
Hematol Educ Program. 2016;(1):485–494.
2015;33:3459–3466.
[66] Tan D, Kim K, Kim JS, et al. The impact of upfront
[78] Palumbo A, Bringhen S, Kumar SK, et al. Second pri-
versus sequential use of bortezomib among patients
mary malignancies with lenalidomide therapy for
with newly diagnosed multiple myeloma (MM): a
newly diagnosed myeloma: a meta-analysis of indi-
joint analysis of the Singapore MM Study Group and
vidual patient data. Lancet Oncol. 2014;15:333–342.
the Korean MM Working Party for the Asian mye-
[79] Zweegman S, van der Hold B, Mellqvist UH, et al.
loma network. Leukemia Res. 2013;37:1070–1076.
[67] Palumbo A, Bringhen S, Mateos M-V, et al. Geriatric Randomized phase III trial in non-transplant eligible
assessment predicts survival and toxicities in elderly patients with newly diagnosed symptomatic multiple
myeloma patients: an International Myeloma myeloma comparing melphalan-prednisonethalido-
Working Group report. Blood 2015;125:2068–2074. mide followed by thalidomide maintenance (MPT-T)
[68] Engelhardt M, Dold SM, Ihorst G, et al. Geriatric versus melphalan-prednisone-lenalidomide followed
assessment in multiple myeloma patients: validation by maintenance with lenalidomide (MPR-R); a joint
of the International Myeloma Working Group (IMWG) study of the Dutch-Belgian Cooperative Trial Group
score and comparison with other common comor- for Hematology Oncology (HOVON) and the Nordic
bidity scores. Haematologica 2016;101:1110–1119. Myeloma Study Group (NMSG). Blood 2014;124:179.
[69] Rose JH, O’Toole EE, Dawson NV, et al. Perspectives, [80] Stewart AK, Jacobus SJ, Fonseca R, et al. E1A06: a
preferences, care practices, and outcomes among phase III trial comparing melphalan, prednisone, and
older and middle-aged patients with late-stage can- thalidomide (MPT) versus melphalan, prednisone,
cer. J Clin Oncol. 2004;22:4907–4917. and lenalidomide (MPR) in newly diagnosed multiple
[70] Durie BG, Hoering A, Abidi MH, et al. Bortezomib myeloma (MM). J Clin Oncol. 2014;32:8511.
with lenalidomide and dexamethasone versus lenali- [81] Laubach J, Garderet L, Mahindra A, et al.
domide and dexamethasone alone in patients with Management of relapsed multiple myeloma: recom-
newly diagnosed myeloma without intent for imme- mendations of the International Myeloma Working
diate autologous stem-cell transplant (SWOG S0777): Group. Leukemia 2016;30:1005–1017.
a randomised, open-label, phase 3 trial. Lancet [82] Avet-Loiseau H, Attal M, Campion L, et al. Long-term
2017;389:519–527. analysis of the IFM 99 trials for myeloma: cytogenetic
[71] Kapoor P, Rajkumar SV, Dispenzieri A, et al. abnormalities [t(4;14), del(17p), 1q gains] play a
Melphalan and prednisone versus melphalan, pred- major role in defining long-term survival. J Clin
nisone and thalidomide for elderly and/or transplant Oncol. 2012;30:1949–1952.
ineligible patients with multiple myeloma: a meta- [83] Sonneveld P, Verelst SG, Lewis P, et al. Review of
analysis. Leukemia 2011;25:689–696. health-related quality of life data in multiple
RESOURCE-STRATIFIED MANAGEMENT OF MYELOMA 13

myeloma patients treated with novel agents. in Lancet Oncol. 2015;16(1):e6]. Lancet Oncol.
Leukemia 2013;27:1959–1969. 2014;15:1195–1206.
[84] Lacy MQ, Hayman SR, Gertz MA, et al. Pomalidomide [95] Barlogie B, Anaissie E, van Rhee F, et al.
(CC4047) plus low dose dexamethasone (Pom/dex) is Incorporating bortezomib into upfront treatment for
active and well tolerated in lenalidomide refractory multiple myeloma: early results of total therapy 3. Br
multiple myeloma (MM). Leukemia 2010;24: J Haematol. 2007;138:176–185.
1934–1939. [96] Dingli D, Arendt BK, Bajzer Z, et al. Evolutionary
[85] Berenson JR, Hilger JD, Yellin O, et al. Replacement dynamics of two related malignant plasma cell lines.
of bortezomib with carfilzomib for multiple myeloma Cell Cycle. 2010;9:3792–3797.
patients progressing from bortezomib combination [97] OECD/WHO (2014). Health expenditure per capita
therapy. Leukemia 2014;28:1529–1536. and in relation to GDP. In: Health at a Glance: Asia/
[86] Rossi A, Mark T, Jayabalan D, et al. BiRd (clarithromy- Pacific 2014: Measuring Progress towards Universal
cin, lenalidomide, dexamethasone): an update on Health Coverage. Paris: OECD Publishing. Health at a
long-term lenalidomide therapy in previously glance: Asia pacific region. WHO 2016.
untreated patients with multiple myeloma. Blood [98] Kanavos P, Vandoros S, Garcia-Gonzalez P. Benefits
2013;121:1982–1985. of global partnerships to facilitate access to medi-
[87] Ghosh N, Tucker N, Zahurak M, et al. Clarithromycin cines in developing countries: a multi-country ana-
overcomes resistance to lenalidomide and dexa- lysis of patients and patient outcomes in GIPAP.
methasone in multiple myeloma. Am J Hematol Global Health. 2009;5:19.
2014;89:E116–E120. [99] Kumar L, Sahoo R. Management of multiple mye-
[88] Cook G, Ashcroft AJ, Cairns DA, et al. National loma in resource-constrained settings. Semin Oncol.
Cancer Research Institute Haemato-oncology Clinical 2016;43:690–694.
Studies Group. The effect of salvage autologous [100] Lopes G, de L. Cost comparison and economic impli-
stem-cell transplantation on overall survival in cations of commonly used originator and generic
patients with relapsed multiple myeloma (final chemotherapy drugs in India. Ann Oncol.
results from BSBMT/UKMF Myeloma X Relapse 2013;24:v13–v16.
[Intensive]): a randomised, open-label, phase 3 trial. [101] Mookerjee A, Gupta R, Jasrotia S, et al. Bortezomib,
Lancet Haematol. 2016;3:e340–e351. Lenalidomide and low-dose dexamethasone (VRD)
[89] Dimopoulos M, Spencer A, Attal M, et al. Multiple versus Lenalidomide and low-dose dexamethasone
Myeloma (010) Study Investigators. Lenalidomide (Ld) for newly-diagnosed Multiple myeloma – A
plus dexamethasone for relapsed or refractory mul- Randomized phase III study-Interim Results. Abstract
tiple myeloma [published correction appears in N # OP-009. Presented at the 16th International
Engl J Med. 2009;361(5): 544]. N Engl J Med. Myeloma Workshop; New Delhi, India; 2017.
2007;357:2123–2132. [102] Yanamandra U, Khattry N, Kumar S, et al. Consensus
[90] Dimopoulos MA, Moreau P, Palumbo A, et al. in the management of multiple myeloma in india at
ENDEAVOR investigators. Carfilzomib and dexa- myeloma state of the art 2016 conference. Indian J
methasone versus bortezomib and dexamethasone Hematol Blood Transfus. 2017;33:15–21.
for patients with relapsed or refractory multiple mye- [103] Evens AM, Jovanovic BD, Su YC, et al. Rituximab-
loma (ENDEAVOR): a randomised, phase 3, open- associated hepatitis B virus (HBV) reactivation in lym-
label, multicentre study. Lancet Oncol. 2016;17: phoproliferative diseases: meta-analysis and examin-
27–38. ation of FDA safety reports. Ann Oncol.
[91] Palumbo A, Chanan-Khan A, Weisel K, et al. 2011;22:1170–1180.
Daratumumab, bortezomib, and dexamethasone for [104] Yeo W, Chan TC, Leung NW, et al. Hepatitis B virus
multiple myeloma. N Engl J Med. 2016;375:754–766. reactivation in lymphoma patients with prior
[92] Dimopoulos AM, Oriol A, Nahi H, et al. resolved hepatitis B undergoing anticancer therapy
Daratumumab, lenalidomide and dexamethasone for with or without rituximab. J Clin Oncol.
multiple myeloma. N Engl J Med. 2016;375: 2009;27:605–611.
1319–1331. [105] Huang YH, Hsiao LT, Hong YC, et al. Randomized
[93] Lonial S, Dimopoulos M, Palumbo A, et al. controlled trial of entecavir prophylaxis for rituxi-
Elotuzumab therapy for relapsed or refractory mul- mab-associated hepatitis B virus reactivation in
tiple myeloma. N Engl J Med. 2015;373:621–631. patients with lymphoma and resolved hepatitis B.
[94] San-Miguel JF, Hungria VT, Yoon SS, et al. J Clin Oncol. 2013;31:2765–2772.
Panobinostat plus bortezomib and dexamethasone [106] Kim IK, Kim BG, Kim W, et al. Clinical prediction of
versus placebo plus bortezomib and dexamethasone failure of lamivudine prophylaxis for hepatitis B
in patients with relapsed or relapsed and refractory virus-infected patients undergoing cytotoxic chemo-
multiple myeloma: a multicentre, randomised, dou- therapy for malignancy. Antimicrob Agents
ble-blind phase 3 trial [published correction appears Chemother. 2012;56:5511–5519.

You might also like