Is This the Time to Introduce Minimal Residual Disease in Multiple ...

33
1 Is This the Time to Introduce Minimal Residual Disease in Multiple Myeloma Clinical Practice? Bruno Paiva 1 , Noemi Puig 2 , Ramón García-Sanz 2 , and Jesús F. San Miguel 1 ; on behalf of the Grupo Español de Mieloma (GEM)/Programa para el Estudio de la Terapéutica en Hemopatías Malignas (PETHEMA) cooperative study groups 1 Clinica Universidad de Navarra, Centro de Investigacion Medica Aplicada (CIMA), Pamplona, 2 Hospital Universitario de Salamanca, Instituto de Investigaion Biomedica de Salamanca, IBMCC (USAL-CSIC), Salamanca, Spain Corresponding Author: Jesús F. San Miguel, Clinica Universidad de Navarra, Centro de Investigacion Medica Aplicada (CIMA), Av. Pio XII 36, 31008 Pamplona, Spain; E-mail: [email protected] Running Title: State of the Art: MRD Testing for Myeloma Disclosure of Potential Conflicts of Interest B. Paiva reports receiving speakers bureau honoraria from Becton, Dickinson and Company, Binding Site, Celgene, Janssen, and Millennium Pharmaceuticals. J.F. San Miguel is a consultant/advisory board member for Bristol-Myers Squibb, Celgene, Janssen, Merck, Millennium Pharmaceuticals, Novartis, and Onyx Pharmaceuticals. No potential conflicts of interest were disclosed by the other authors. Research. on April 13, 2018. © 2015 American Association for Cancer clincancerres.aacrjournals.org Downloaded from Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on March 9, 2015; DOI: 10.1158/1078-0432.CCR-14-2841

Transcript of Is This the Time to Introduce Minimal Residual Disease in Multiple ...

Page 1: Is This the Time to Introduce Minimal Residual Disease in Multiple ...

1

Is This the Time to Introduce Minimal Residual Disease in Multiple

Myeloma Clinical Practice?

Bruno Paiva1, Noemi Puig2, Ramón García-Sanz2, and Jesús F. San Miguel1;

on behalf of the Grupo Español de Mieloma (GEM)/Programa para el Estudio

de la Terapéutica en Hemopatías Malignas (PETHEMA) cooperative study

groups

1Clinica Universidad de Navarra, Centro de Investigacion Medica Aplicada

(CIMA), Pamplona, 2Hospital Universitario de Salamanca, Instituto de

Investigaion Biomedica de Salamanca, IBMCC (USAL-CSIC), Salamanca,

Spain

Corresponding Author: Jesús F. San Miguel, Clinica Universidad de Navarra,

Centro de Investigacion Medica Aplicada (CIMA), Av. Pio XII 36, 31008

Pamplona, Spain; E-mail: [email protected]

Running Title: State of the Art: MRD Testing for Myeloma

Disclosure of Potential Conflicts of Interest

B. Paiva reports receiving speakers bureau honoraria from Becton, Dickinson

and Company, Binding Site, Celgene, Janssen, and Millennium

Pharmaceuticals. J.F. San Miguel is a consultant/advisory board member for

Bristol-Myers Squibb, Celgene, Janssen, Merck, Millennium Pharmaceuticals,

Novartis, and Onyx Pharmaceuticals. No potential conflicts of interest were

disclosed by the other authors.

Research. on April 13, 2018. © 2015 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on March 9, 2015; DOI: 10.1158/1078-0432.CCR-14-2841

Page 2: Is This the Time to Introduce Minimal Residual Disease in Multiple ...

2

Abstract

Increasing therapeutic options and prolonged survival in multiple myeloma (MM)

have raised interest on the concept of depth of response and its importance to

predict patients’ outcome. While the efficacy of current treatment approaches

has greatly improved in the last decade, the definition of complete response

(CR) remains unaltered and continues to use conventional serological and

morphological techniques. That notwithstanding, there is growing interest in

minimal residual disease (MRD) monitoring, which has emerged in recent years

as one of the most relevant prognostic factors in MM. MRD can be assessed

both inside (e.g., immunophenotypic and molecular techniques) and outside the

bone marrow (e.g., PET-CT). Here, we focus on flow- and molecular-based

assays by which different cooperative groups have demonstrated the efficacy of

MRD assessment to predict outcomes even among patients in CR, and

irrespectively of disease risk. Although, further standardization is still required,

the time has come to implement MRD monitoring in prospective clinical trials as

a sensitive tool to evaluate treatment efficacy and for risk-adapted treatment,

particularly in the consolidation and maintenance settings. Here we present a

comprehensive and critical review on the methodological aspects, specific

characteristics, as well as clinical significance of MRD monitoring by flow

cytometry, PCR and next generation sequencing.

Research. on April 13, 2018. © 2015 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on March 9, 2015; DOI: 10.1158/1078-0432.CCR-14-2841

Page 3: Is This the Time to Introduce Minimal Residual Disease in Multiple ...

3

Introduction

In the last decades, important changes have been introduced in the treatment of

multiple myeloma (MM) patients, resulting in unprecedented complete remission

(CR) rates and prolonged progression-free (PFS) and overall survival (OS) (1-

3). In 2006 the International Myeloma Working Group proposed a new CR

definition, and introduced the normalization of serum free light-chains (sFLC)

and absence of bone marrow (BM) clonal plasma cells (PCs) by

immunohistochemistry/immunofluorescence as additional requirements to

define stringent CR (4). Recently, Kapoor et al. (5) demonstrated the added

prognostic value of the stringent over conventional CR criteria, even though

other groups have failed to show additional value of sFLC assessment among

CR patients (6, 7). However, the sensitivity of

immunohistochemistry/immunofluorescence is rather low (10-2) due to the

recovery of normal PCs after therapy that normalize kappa/lambda ratios (8).

Thus, it becomes evident that current criteria used to assess response lag

behind the extraordinary evolution in the treatment of MM patients, and more

sensitive techniques are needed to detect true minimal residual disease (MRD)

for new CR definitions. This would contribute to improve monitoring of treatment

efficacy as well as to avoid over and under treatment, particularly during

consolidation and maintenance.

Current techniques to monitor MRD can be divided into those measuring

extramedullary vs. those detecting intramedullary disease. Magnetic resonance

imaging (MRI) and 18-fluoro-deoxyglucose positron emission tomography-

computerized tomography (PET/CT) have emerged as promising tools to

measure extramedullary MRD. In particular, PET/CT has shown to be of

Research. on April 13, 2018. © 2015 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on March 9, 2015; DOI: 10.1158/1078-0432.CCR-14-2841

Page 4: Is This the Time to Introduce Minimal Residual Disease in Multiple ...

4

prognostic value as soon as at day 7 of induction therapy (9) and more

importantly, among patients in conventional CR after HDT/ASCT (10). For

detection of intramedullary disease, both multiparameter flow cytometry

immunophenotyping and molecular assessment of immunoglobulin

rearrangements are the pivotal techniques. The present review focus on current

state-of-the-art intramedullary MRD monitoring in MM.

Methodical Characterization of MRD Techniques

Multiparameter flow cytometry (MFC)

Simultaneous assessment of CD38 and CD138 represents the best marker

combination for specific identification of PC (11-14). In the event of anti-CD38

and/or anti-CD138 therapies, other antigens such as CD229, CD319 or CD54

could be alternative markers for PC identification. Phenotypically aberrant clonal

PCs typically show: i) underexpression of CD19, CD27, CD38, and/or CD45; ii)

overexpression of CD56; iii) asynchronous expression of CD117 (15-24).

Accordingly, these antigens are consensually used for the clinical study of BM

PCs (25) and at least one (8-color) combination of such markers is highly

recommended for MM flow-MRD monitoring. No single parameter reliably

distinguishes clonal from normal PCs; however, a multiparameter approach that

evaluates all markers used in a single tube can readily identify PC aberrant

phenotypes, provided a sufficient number of cellular events are evaluated in the

flow cytometer (26).

Flow-based MRD monitoring has benefited from polychromatic

cytometers that allow simultaneous assessment of ≥8-markers at the single-cell

level which, coupled with novel software, results in more accurate discrimination

of clonal vs. normal PCs (27). The principal component analysis (PCA) of

Research. on April 13, 2018. © 2015 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on March 9, 2015; DOI: 10.1158/1078-0432.CCR-14-2841

Page 5: Is This the Time to Introduce Minimal Residual Disease in Multiple ...

5

unified data files allows automatic multidimensional separation of all cellular

clusters present in a sample. PCA-based interpretation of flow cytometric data

facilitates the development of normal and tumor reference libraries, automated

separation of populations within a sample, and prospective detection and

tracking of any aberrant cell population. Such strategy is likely the method of

choice for accurate and semi-automated flow-MRD monitoring.

Allele specific oligonucleotide polymerase chain reaction (ASO-PCR)

PCR-based detection of MM MRD relies on the identification of persistent tumor

cells through the amplification of the immunoglobulin heavy-chain (IGHV) gene

rearrangement (28). Consensus primers binding to the less variable regions of

the IGHV rearrangement (FR o framework) were initially used for the

identification of residual disease. This approach amplifies the monoclonal IGHV

rearrangement but also those from normal B-cells and thus, the sensitivity

reached (10-1 - 10-2) was insufficient for MRD detection. Afterwards, specific

primers (ASO: allelic specific oligonucleotide) complementary to the most

variable regions of the IGVH rearrangement (CDRs or complementary

determinant regions) were used. The limit of detection with this approach was

optimal (10-4-10-6) but the results obtained were merely qualitative (29, 30). The

subsequent step was the implementation of RQ-PCR strategies that allow the

exact quantification of tumor cells in real-time. This method is based on the use

of an ASO primer (usually complementary to the CDR3 region) together with a

consensus primer (frequently complementary to the JH region) and a

fluorescent probe to monitor the amplification.

Next generation sequencing (NGS)

Research. on April 13, 2018. © 2015 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on March 9, 2015; DOI: 10.1158/1078-0432.CCR-14-2841

Page 6: Is This the Time to Introduce Minimal Residual Disease in Multiple ...

6

In recent years, sequencing technologies that quickly perform millions of reads

of DNA fragments have emerged. This technology not only allows to distinguish

normal from tumor DNA, but also to detect previously known tumor-specific

sequences within normal DNA fragments (i.e., MRD monitoring). Current NGS

methods include: i) pyrosequencing, based on the luminometric detection of the

pyrophosphate released when individual nucleotides are added to DNA

templates from an emulsion PCR; ii) multiplex sequencing-by-synthesis

technology, that rely on light signals emitted during the re-synthesis of small

DNA fragments previously produced by bridge amplification; and iii) ion

semiconductor sequencing, that detects hydrogen ions liberated during DNA

polymerization Using these techniques rearranged B-cell receptor (BCR) and T-

cell receptor (TCR) genes can be deeply sequenced (31-35). These methods

use a consensus PCR to amplify all possible BCR or TCR rearrangements

which, at diagnosis, allow identifying and sequencing monoclonal

rearrangements (35). After therapy, monoclonal rearrangements can be

investigated among thousands of normal cells through several millions reads,

providing high specificity and sensitivity for MRD detection of BCR and TCR

genes.

Advantages and Disadvantages of MRD Techniques

MFC

An increasingly higher number of flow cytometry laboratories use digital

instruments that provide simultaneous assessment of more parameters per tube

(≥8), as well as a superior event acquisition and analysis of a greater cell

numbers than previously seen with 4-color instruments. The availability of digital

cytometers coupled with novel sample preparation methods allow for fast- and

Research. on April 13, 2018. © 2015 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on March 9, 2015; DOI: 10.1158/1078-0432.CCR-14-2841

Page 7: Is This the Time to Introduce Minimal Residual Disease in Multiple ...

7

cost-effective measurement of millions of leukocytes. Consequently, while

previous MFC studies defined MRD as the presence of a discrete population of

clonal PCs equal or above the 0.01% limit of detection, nowadays such

threshold stands at 0.001% (10-5). Because clonal PCs are readily distinguished

from normal PCs on the basis of their aberrant phenotypes, flow-MRD is

applicable to virtually all patients and does not require patients’ diagnostic

phenotypic profile. Furthermore, the flow-MRD assays incorporates a quality

check of the whole sample cellularity (i.e., B-cell precursors, erythroblasts, etc.),

which is critical to ensure sample quality since hemodiluted BM aspirates can

lead to false-negative results. Even though hemodilution can be assessed with

a microscope - providing that the sample in which hemodilution is being

assessed is exactly the same than that where MRD will be assessed -, the

possibility for MFC to simultaneously analyze MRD and hemodilution

automatically in the same sample is more accurate and particularly attractive.

Extensive expertise in MFC analysis requirement and the lack of a well-

standardized flow-MRD method have been pointed as important disadvantages

of MFC immunophenotyping. However, automated identification and

characterization of cell populations, coupled with reference databases in the

context of full technical standardization have now emerged as the probable

solution for these problems. There is now the unmet need for different MFC

groups to adopt single, standardized and validated antibody panels, sample

processing and cell-analysis methods such as those being developed by the

EuroFlow consortium for MFC to become a universal and fully standardized

option for MRD assessment. Another potential limitation of MFC could be to

ignore potential MM cancer stem cells outside the PC compartment (i.e.,

Research. on April 13, 2018. © 2015 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on March 9, 2015; DOI: 10.1158/1078-0432.CCR-14-2841

Page 8: Is This the Time to Introduce Minimal Residual Disease in Multiple ...

8

memory B-cells) (36). However, recent investigations conducted with optimized

molecular assessment of clonal VDJ sequences among FACS-sorted peripheral

blood B-cell subsets revealed that such clonotypic cells are either absent, or

present below the detection limits (10-6) (37).

Regarding the potential impact of genetic heterogeneity and clonal tiding

after treatment on the feasibility of MFC to detect MRD, it should be highlighted

that the multidimensional approach of current flow cytometry

immunophenotyping not only allows to detect clonal heterogeneity in

approximately 30% of newly-diagnosed patients, but also to monitor all different

phenotypic sub-clones throughout patients’ treatment, thereby assessing

potential (phenotypical) clonal selection upon therapy. Nevertheless, it should

be noted that according to the experience of the Medical Research Council

(MRC) and the Grupo Español de Mieloma (GEM) groups based on large

patient cohorts, there are no major antigenic shifts for consensus markers (e.g.,

CD19, CD38, CD45, or CD56) used to monitor MRD (GEM; unpublished data).

Accordingly, potential clonal evolution throughout the course of treatment does

not influence the efficacy of MFC-based MRD assessment.

ASO-PCR

PCR approaches for MRD detection do not require an immediate sample

processing since they are unaffected by pre-analytical biases such as loss of

viable cells over time (38, 39). Furthermore, the MRD target used is based on

the uniqueness of clonal IGHV rearrangements which leads to a high sensitivity,

adequate for MRD detection (10-5 -10-6) (40, 41). PCR strategies have passed

an exhaustive process of validation and standardization for MRD testing in

different hematological malignancies, which make them readily available and

Research. on April 13, 2018. © 2015 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on March 9, 2015; DOI: 10.1158/1078-0432.CCR-14-2841

Page 9: Is This the Time to Introduce Minimal Residual Disease in Multiple ...

9

reproducible among different centers (42, 43). However, these approaches

require diagnostic samples with high tumor load to identify patient-specific

clonotypic sequences (44, 45). Moreover, MM is a post-germinal center

neoplasia characterized by a high rate of somatic hypermutations both in the

heavy- and light-chain immunoglobulin genes (46-48). Such mutations prevent

the annealing of consensus primers, limit clonal detection, sequencing success

rate and overall ASO performance, forcing the use of specific primers/probes

instead (49). Conversely, it has been recently reported that the use of CD138+

positively selected BM PCs may significantly increase the applicability of PCR-

based MRD studies in MM (50). Nevertheless, the technique remains costly,

laborious, and accordingly, difficult to implement into routine clinical practice.

NGS

The greatest advantage of NGS approaches for MRD detection in MM is its

sensitivity which, without compromising specificity, is estimated to be in the

range of 10-5-10-6 (35, 51). Moreover, NGS can identify both stable and dynamic

aspects of the BCR rearrangement, including potential clonal tiding (52).

However, the presence of subclonality in diagnostic samples is typically below

7% of all patients, and virtually impossible to be distinguished from biallellic

rearrangements (53). Furthermore, the main clonal rearrangement is usually

stable from diagnosis to relapse (B. Puig et al.; submitted for publication).

Therefore, since clonal Ig gene sequences remain stable and because current

MRD monitoring by ASO-PCR or NGS does not focus on gene mutations or

copy number abnormalities, molecular techniques are not influenced by genetic

heterogeneity and clonal tiding throughout patients’ treatment. Other

advantages that NGS offers are superior sensitivity, potential scalability and

Research. on April 13, 2018. © 2015 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on March 9, 2015; DOI: 10.1158/1078-0432.CCR-14-2841

Page 10: Is This the Time to Introduce Minimal Residual Disease in Multiple ...

10

less methodological complexity. In particular, the latest advantage would be

crucial in MM since it removes the need of standard curve construction, which is

the main reason of ASO-PCR failure in MM (49). However, there are also some

disadvantages. Molecular-based approaches cannot distinguish hemodiluted

from good quality samples. Albeit the applicability of NGS is superior to that of

ASO-PCR, still 10% of patients will be missed during the initial PCR step (51).

In addition, MRD quantitation is only approximate, because the efficacy of

amplification is highly variable depending on the specific sequence of the

rearrangement (54). Since only B-cells have rearranged immunoglobulins,

NGS-based MRD monitoring will only quantify B-cells; accordingly, global MRD

quantitation requires the addition of an artificial internal control for such

quantification. Finally, NGS is a labor-intensive and expensive technology, and

it is yet not commonly available for clinical practice.

Clinical Results with (Intramedullary) MRD Techniques

MFC

The prognostic value of MFC-based MRD monitoring in MM was introduced in

2002 by San Miguel (55) and Rawstron (20); both studies suggesting the utility

of monitoring the BM PC compartment among MM patients treated with

conventional or high-dose chemotherapy, even if such patients were in CR (20).

This initial positive experience led the Spanish and UK groups to implement

their corresponding 4- and 6-color flow-MRD methods in large clinical trials. In

the PETHEMA/GEM2000 study, flow-MRD was identified as the most relevant

prognostic factor in a series of 295 newly-diagnosed MM patients receiving

uniform treatment including HDT/SCT (56). MRD negativity at day 100 after

ASCT translated to significantly improved PFS and OS, and the impact of MRD

Research. on April 13, 2018. © 2015 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on March 9, 2015; DOI: 10.1158/1078-0432.CCR-14-2841

Page 11: Is This the Time to Introduce Minimal Residual Disease in Multiple ...

11

was equally relevant among patients in CR. Similarly, in the intensive-pathway

of the MRC Myeloma IX study, MRD-negativity at day 100 after ASCT was

predictive of favorable PFS and OS (57). This outcome advantage was equally

demonstrable in patients achieving CR. Since in MM there has been extensive

debate on whether attaining deep levels of remission (i.e., CR) would be critical

to all patients or in turn is particularly relevant for patients with high-risk disease,

it is important to emphasize that both the PETHEMA/GEM and UK groups have

demonstrated that risk assessment by FISH and flow-MRD monitoring were of

independent prognostic value in transplant-eligible patients (56, 57).

Furthermore, it is particularly interesting to observe the benefit of achieving

MRD-negativity in high-risk patients, whose outcome becomes similar to that of

standard-risk patients (25). Accordingly, further research on the role of MRD as

a surrogate for prolonged OS among high-risk patients is warranted, since it

could represent an attractive clinical end-point to improve the overall poor

prognosis of this patient population. Thus, combined cytogenetic/FISH

evaluation at diagnosis plus MRD assessment after HDT/ASCT (day +100),

provided powerful risk stratification, which also resulted in a highly-effective

approach to identify patients with unsustained CR and dismal outcomes (25).

Collectively, these results confirm the superiority of MRD assessment over

conventional response criteria to predict outcome in distinct MM genetic

subgroups. The effect of maintenance therapy with thalidomide was also

assessed in the UK study. Interestingly, MRD-positive patients randomized to

the maintenance arm experienced significantly prolonged PFS as compared to

the placebo arm; in MRD-negative patients a similar trend was observed (57).

Further analyses by the PETHEMA/GEM have shown that combining the

Research. on April 13, 2018. © 2015 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on March 9, 2015; DOI: 10.1158/1078-0432.CCR-14-2841

Page 12: Is This the Time to Introduce Minimal Residual Disease in Multiple ...

12

prognostic information of cytogenetics at diagnosis plus MRD assessment at

day 100 after HDT/ASCT resulted in a highly effective approach to identify

patients with unsustained CR and dismal outcomes (2-years median OS): those

with baseline high-risk cytogenetics plus persistent MRD after ASCT (58). The

Spanish myeloma group has also shown that it was possible for elderly patients

treated with bortezomib-based induction regimens to achieve MRD-negativity,

and that flow-MRD resulted in superior patient prognostication than

conventional and stringent CR response criteria (7). More recently, the

Intergroupe Francophone du Myélome has reported on the prognostic value of

their 7-color flow-MRD method implemented in a recent phase II study (59).

Overall, 68% of patients achieved MRD negativity and none of these patients

relapsed. Thus, it is plausible to assume that albeit the already well-established

link between patients’ flow-MRD status and survival, the true clinical utility of

flow-based MRD assessment is likely to be significantly improved with more

sensitive (10-5) and multidimensional (≥8-colors) immunophenotyping. Table 1

summarizes the most relevant flow-based MRD studies reported in MM.

ASO-PCR

A considerable number of studies have explored the value of PCR-based MRD

monitoring in MM (Table 2) (60-66). Although initial observations lacked clinical

value, additional studies performed in patients undergoing autologous or

allogeneic SCT unraveled the prognostic value of reaching molecular

remissions (Table 2) (39-41, 49, 50, 65, 67-72) Using non-quantitative

approaches, the percentage of molecular remissions observed after allogeneic

SCT was significantly higher as compared to patients undergoing autologous

SCT, suggesting a role for this technique to evaluate treatment efficacy.

Research. on April 13, 2018. © 2015 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on March 9, 2015; DOI: 10.1158/1078-0432.CCR-14-2841

Page 13: Is This the Time to Introduce Minimal Residual Disease in Multiple ...

13

Furthermore, Lipinski et al., in a retrospective study performed in 13 patients

undergoing ASCT suggested the potential value of ASO-PCR monitoring to

predict progression (73), and this notion of MRD reappearance heralding

relapse has been recently confirmed by the GIMEMA group (72).

Semi-quantitative and quantitative approaches have also been used to

predict patients’ outcome according to MRD levels. Korthals et al. in a cohort of

53 patients undergoing ASCT have shown that different MRD levels by ASO-

RQ-PCR before ASCT allowed two discriminate two groups of patients with

different PFS and OS (0,2% 2IgH/βactin) (41). Putkonen et al. in a series of 37

patients undergoing autologous and allogeneic stem cell transplantation defined

0.01% as the optimal MRD threshold to distinguish two groups of patients with

different PFS and also OS (71). Puig et al., in a recent study that included 103

patients undergoing ASCT also found 10-4 as the most significant cutoff level,

distinguishing two subgroups with different PFS and, when applied to patients in

conventional CR, also different OS (50). Finally, Ladetto et al. with nested and

ASO-RQ-PCR have reported on the significant reduction of residual tumor load

after bortezomib, thalidomide and dexamethasone (VTD) consolidation, which

translated into prolonged PFS (70). A recent update of the study showed that

MRD monitoring also predicted for different OS: 72% at 8 years for patients in

major MRD response vs. 48% for those with positive MRD (72).

NGS

Since NGS-based MRD monitoring is still a relatively recent approach, there is

yet few data in MM. However, the PETHEMA/GEM has already described

favorable and promising results in a series of 133 MM patients including both

transplant and non-transplant eligible cases. The applicability of NGS-based

Research. on April 13, 2018. © 2015 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on March 9, 2015; DOI: 10.1158/1078-0432.CCR-14-2841

Page 14: Is This the Time to Introduce Minimal Residual Disease in Multiple ...

14

MRD monitoring using the LymphoSIGHT® methodology was of 90%. The

median TTP and OS of MRD-negative cases were of 80 months and not

reached, respectively (51). Importantly, Martinez-Lopez identified three groups

of patients with different TTP: patients with high (<10-3), intermediate (10-3 to 10-

5), and low (>10-5) MRD levels showed significantly different TTP: 27, 48, and

80 months, respectively, which indicates that the deepest the quality of CR, the

better the patients outcome (51). Similar results are also now being observed

with more sensitive MFC (GEM; unpublished observations).

Discordant Results between (Intramedullary) MRD Techniques

MFC vs. ASO-PCR

Three studies have directly compared MFC and ASO-PCR as alternative

methods for MRD detection in MM. In 2005, Sarasquete et al. quantified MRD

levels in 24 patients at day 100 after HDT/ASCT using both techniques, and

observed discordant results in 6 out of the 24 cases; all of them negative by

MFC but positive by ASO-RQ-PCR (39). Discordances were attributed to the

limit of MRD detection by each technique, since the median number of clonal

PCs in PCR+MFC- cases was 0.014%. A second study by Lioznov et al.,

performed on 69 samples from 13 patients undergoing allogeneic SCT, found a

very high correlation between both techniques. Virtually all results were

concordant, and both techniques had similar applicability and prognostic value

(74). More recently, Puig et al. compared the results of MRD assessment by

MFC and ASO-PCR in 103 cases undergoing HDT/ASCT, and only observed

18 discordant results (11 cases were PCR+MFC- while the other 7 were MFC+

but PCR-). No differences in survival were observed between the discordant

groups of patients (49).

Research. on April 13, 2018. © 2015 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on March 9, 2015; DOI: 10.1158/1078-0432.CCR-14-2841

Page 15: Is This the Time to Introduce Minimal Residual Disease in Multiple ...

15

MFC vs. NGS

Only one study has compared MFC vs. NGS for MRD monitoring in MM (51).

From a total of 99 patients, 82 had concordant results (60 double-positive and

22 double-negative), twelve were MFC–NGS+ and five were MFC+NGS–.

Discordances are likely related to differences in sensitivity and specificity, which

theoretically favor NGS over 4-color MFC. However, it cannot be excluded that

certain clonal rearrangements could be under-amplified against polyclonal

rearrangements and, if present in very low numbers (typical situation in MRD

samples), these could remain undetectable. Noteworthy, the time-to

progression of MRD- patients by NGS was slightly better than NGS+MFC- cases

(P = .05). However, this study was a retrospective comparison and the MFC

approach was a conventional 4-color staining of a limited number of cells (10-

fold less compared to current MFC studies) (51).

ASO-PCR vs. NGS

Only two studies have compared ASO-PCR vs. NGS. Ladetto et al. has

reported preliminary data on 10 MM patients; 8 were evaluable by RQ-PCR, 8

by NGS and 6 by both methods (75). Among total follow-up samples, 20

discordances were recorded: 12 qualitative and 8 quantitative discordances. In

9 samples, RQ-PCR yielded a positive or 1 log higher result compared to NGS,

while the opposite occurred in 11 cases. Overall, major discordance rates in

MM were comparable to ALL and MCL, while minor and quantitative

discordances appeared slightly more frequent (75). Martinez-Lopez et al.

performed a similar comparison in 46 patients, and observed a concordance

rate of 85% (51). Thus, further studies are warranted to elucidate the clinical

Research. on April 13, 2018. © 2015 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on March 9, 2015; DOI: 10.1158/1078-0432.CCR-14-2841

Page 16: Is This the Time to Introduce Minimal Residual Disease in Multiple ...

16

significance of discordant results from two molecular techniques that, albeit

methodologically different, have similar sensitivity.

The ideal MRD test should fulfill a minimum of several relevant

characteristics: i) high applicability, ii) high sensitivity, iii) readiness and rapid

turnaround, iv) feasible in low sample volumes, v) reproducibility, and, vi) of

clinical utility. Because the cost of MRD assessment is significantly lower as

compared to the cost of some drugs its importance is less relevant; however, in

the event of long-term follow-up sequential analysis (similarly to what is done in

CML or ALL), total cost becomes significantly higher and therefore important to

evaluate. While we wait for further results on the comparison between ASO-

PCR and NGS, the higher applicability of the latter would favor its incorporation

in clinical trials. Unfortunately, data on prospective comparison between the

next-generation MFC vs. NGS is not yet available, and knowledge on head-to-

head applicability and sensitivity are missing. Altogether, this precludes us to

indicate at present, a favorite methodology to be implemented in clinical trials.

For routine clinical practice, we would support at this moment the use of next-

generation MFC given its wide availability, cost-effectiveness, and

independence of previous collection of a diagnostic sample.

Concluding Remarks and Future Direction

Approximately fifteen years after the first published results, MRD detection has

emerged as one of the most relevant prognostic factors in MM. Because MRD

represents the collective end result of all of the cellular mechanisms that

determine a patient response to a given therapy, MRD assessment affords

prognostic information even among patients in CR, and irrespectively of disease

risk. Accordingly, there is increasing interest on MRD monitoring as a sensitive

Research. on April 13, 2018. © 2015 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on March 9, 2015; DOI: 10.1158/1078-0432.CCR-14-2841

Page 17: Is This the Time to Introduce Minimal Residual Disease in Multiple ...

17

tool to evaluate treatment efficacy, to become a surrogate marker for clinically

relevant end-points, and for risk-adapted treatment, particularly in the

consolidation and maintenance setting. However, the patchy pattern of BM

infiltration, typically observed in MM, leads to a certain degree of uncertainty

regarding an MRD-negative result irrespectively of the technique adopted: does

it represent real absence of clonal PCs, or is it due to sampling error? The

possibility of patchy BM infiltration or extramedullary involvement represent a

challenge for both immunophenotypic- and molecular-based MRD detection in

single BM aspirates. This highlights the value of sensitive imaging techniques to

re-define CR both at the intramedullary (e.g., whole body MRI and PET/CT) and

extra-medullary levels (PET/CT). However, standardization of imaging

techniques and comparison with other sensitive BM-based MRD methods are

still lacking. By contrast, the persistence of MRD tumor cells is always an

adverse prognostic feature, even among CR patients, envisioning that for the

time being, it would also be safer to take clinical decisions based on MRD-

positivity than on MRD-negativity. The clinical applicability of MRD in MM has

been prospectively confirmed in two large transplant-based studies (56, 57) and

a relatively smaller (yet prospective) clinical trial on transplant-ineligible MM

patients (7); such results were also retrospectively reproduced on smaller

patient series mostly by using molecular techniques. In all studies, the PFS of

MRD-negative patients at least doubled that of MRD-positive CR patients;

conversely, both MFC and ASO-PCR showed that CR patients with persistent

MRD had significantly inferior OS vs. MRD-negative cases. MDR has also

proven to be relevant in both standard- as well as high-risk patients. Altogether,

these results strongly support the rationale for implementing MRD assessment

Research. on April 13, 2018. © 2015 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on March 9, 2015; DOI: 10.1158/1078-0432.CCR-14-2841

Page 18: Is This the Time to Introduce Minimal Residual Disease in Multiple ...

18

to re-define and improve current CR criteria in MM. The time has come to

implement MRD monitoring in prospective clinical trials and fully establish its

role in the management of MM patients.

Grant Support

This study was supported by the Cooperative Research Thematic Network of

the Red de Cancer (Cancer Network of Excellence) (RD12/0036/0058; to J. San

Miguel); Instituto de Salud Carlos III, Spain, Instituto de Salud Carlos

III/Subdirección General de Investigación Sanitaria (FIS: PI1202311; to R.

Garcia-Sanz; Sara Borrell: CD13/00340; to B. Paiva); Junta de Castilla y León

(HUS412A12-1; to B. Paiva); and Asociación Española Contra el Cáncer

(GCB120981SAN; to J. San Miguel).

References

1. Mateos MV, San Miguel JF. How should we treat newly diagnosed

multiple myeloma patients? Hematology Am Soc Hematol Educ Program

2013;2013:488-95.

2. McCarthy PL, Hahn T. Strategies for induction, autologous hematopoietic

stem cell transplantation, consolidation, and maintenance for transplantation-

eligible multiple myeloma patients. Hematology Am Soc Hematol Educ Program

2013;2013:496-503.

3. Kumar SK, Dispenzieri A, Lacy MQ, Gertz MA, Buadi FK, Pandey S, et

al. Continued improvement in survival in multiple myeloma: changes in early

mortality and outcomes in older patients. Leukemia 2014;28:1122-8.

4. Durie BG, Harousseau JL, Miguel JS, Blade J, Barlogie B, Anderson K,

et al. International uniform response criteria for multiple myeloma. Leukemia

2006;20:1467-73.

Research. on April 13, 2018. © 2015 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on March 9, 2015; DOI: 10.1158/1078-0432.CCR-14-2841

Page 19: Is This the Time to Introduce Minimal Residual Disease in Multiple ...

19

5. Kapoor P, Kumar SK, Dispenzieri A, Lacy MQ, Buadi F, Dingli D, et al.

Importance of achieving stringent complete response after autologous stem-cell

transplantation in multiple myeloma. J Clin Oncol 2013;31:4529-35.

6. de Larrea CF, Cibeira MT, Elena M, Arostegui JI, Rosinol L, Rovira M, et

al. Abnormal serum free light chain ratio in patients with multiple myeloma in

complete remission has strong association with the presence of oligoclonal

bands: implications for stringent complete remission definition. Blood

2009;114:4954-6.

7. Paiva B, Martinez-Lopez J, Vidriales MB, Mateos MV, Montalban MA,

Fernandez-Redondo E, et al. Comparison of immunofixation, serum free light

chain, and immunophenotyping for response evaluation and prognostication in

multiple myeloma. J Clin Oncol 2011;29:1627-33.

8. San-Miguel JF, Paiva B, Gutiérrez NC. New tools for diagnosis and

monitoring of multiple myeloma. Am Soc Clin Oncol Educ Book 2013. doi:

10.1200/EdBook_AM.2013.33.e313.

9. Usmani SZ, Mitchell A, Waheed S, Crowley J, Hoering A, Petty N, et al.

Prognostic implications of serial 18-fluoro-deoxyglucose emission tomography

in multiple myeloma treated with total therapy 3. Blood 2013;121:1819-23.

10. Zamagni E, Patriarca F, Nanni C, Zannetti B, Englaro E, Pezzi A, et al.

Prognostic relevance of 18-F FDG PET/CT in newly diagnosed multiple

myeloma patients treated with up-front autologous transplantation. Blood

2011;118:5989-95.

11. Almeida J, Orfao A, Ocqueteau M, Mateo G, Corral M, Caballero MD, et

al. High-sensitive immunophenotyping and DNA ploidy studies for the

Research. on April 13, 2018. © 2015 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on March 9, 2015; DOI: 10.1158/1078-0432.CCR-14-2841

Page 20: Is This the Time to Introduce Minimal Residual Disease in Multiple ...

20

investigation of minimal residual disease in multiple myeloma. Br J Haematol

1999;107:121-31.

12. Garcia-Sanz R, Orfao A, Gonzalez M, Tabernero MD, Blade J, Moro MJ,

et al. Primary plasma cell leukemia: clinical, immunophenotypic, DNA ploidy,

and cytogenetic characteristics. Blood 1999;93:1032-7.

13. Lin P, Owens R, Tricot G, Wilson CS. Flow cytometric immunophenotypic

analysis of 306 cases of multiple myeloma. Am J Clin Pathol 2004;121:482-8.

14. Rawstron AC, Barrans SL, Blythe D, English A, Richards SJ, Fenton JA,

et al. In multiple myeloma, only a single stage of neoplastic plasma cell

differentiation can be identified by VLA-5 and CD45 expression. Br J Haematol

2001;113:794-802.

15. Dahl IM, Rasmussen T, Kauric G, Husebekk A. Differential expression of

CD56 and CD44 in the evolution of extramedullary myeloma. Br J Haematol

2002;116:273-7.

16. Davies FE, Forsyth PD, Rawstron AC, Owen RG, Pratt G, Evans PA, et

al. The impact of attaining a minimal disease state after high-dose melphalan

and autologous transplantation for multiple myeloma. Br J Haematol

2001;112:814-9.

17. Mateo MG, San MI, Orfao de MA. Immunophenotyping of plasma cells in

multiple myeloma. Methods Mol Med 2005;113:5-24.

18. Nowakowski GS, Witzig TE, Dingli D, Tracz MJ, Gertz MA, Lacy MQ, et

al. Circulating plasma cells detected by flow cytometry as a predictor of survival

in 302 patients with newly diagnosed multiple myeloma. Blood 2005;106:2276-

9.

Research. on April 13, 2018. © 2015 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on March 9, 2015; DOI: 10.1158/1078-0432.CCR-14-2841

Page 21: Is This the Time to Introduce Minimal Residual Disease in Multiple ...

21

19. Perez-Andres M, Almeida J, Martin-Ayuso M, Moro MJ, Martin-Nunez G,

Galende J, et al. Clonal plasma cells from monoclonal gammopathy of

undetermined significance, multiple myeloma and plasma cell leukemia show

different expression profiles of molecules involved in the interaction with the

immunological bone marrow microenvironment. Leukemia 2005;19:449-55.

20. Rawstron AC, Davies FE, DasGupta R, Ashcroft AJ, Patmore R, Drayson

MT, et al. Flow cytometric disease monitoring in multiple myeloma: the

relationship between normal and neoplastic plasma cells predicts outcome after

transplantation. Blood 2002;100:3095-100.

21. Robillard N, Pellat-Deceunynck C, Bataille R. Phenotypic

characterization of the human myeloma cell growth fraction. Blood

2005;105:4845-8.

22. San Miguel JF, Almeida J, Mateo G, Blade J, Lopez-Berges C, Caballero

D, et al. Immunophenotypic evaluation of the plasma cell compartment in

multiple myeloma: a tool for comparing the efficacy of different treatment

strategies and predicting outcome. Blood 2002;99:1853-6.

23. San Miguel JF, Gutierrez NC, Mateo G, Orfao A. Conventional

diagnostics in multiple myeloma. Eur J Cancer 2006;42:1510-9.

24. Moreau P, Robillard N, Jego G, Pellat C, Le Gouill S, Thoumi S, et al.

Lack of CD27 in myeloma delineates different presentation and outcome. Br J

Haematol 2006;132:168-70.

25. Rawstron AC, Orfao A, Beksac M, Bezdickova L, Brooimans RA,

Bumbea H, et al. Report of the European Myeloma Network on multiparametric

flow cytometry in multiple myeloma and related disorders. Haematologica

2008;93:431-8.

Research. on April 13, 2018. © 2015 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on March 9, 2015; DOI: 10.1158/1078-0432.CCR-14-2841

Page 22: Is This the Time to Introduce Minimal Residual Disease in Multiple ...

22

26. Paiva B, Almeida J, Perez-Andres M, Mateo G, Lopez A, Rasillo A, et al.

Utility of flow cytometry immunophenotyping in multiple myeloma and other

clonal plasma cell-related disorders. Cytometry B Clin Cytom 2010;78:239-52.

27. van Dongen JJ, Lhermitte L, Bottcher S, Almeida J, van der Velden VH,

Flores-Montero J, et al. EuroFlow antibody panels for standardized n-

dimensional flow cytometric immunophenotyping of normal, reactive and

malignant leukocytes. Leukemia 2012;26:1908-75.

28. Davies FE, Rawstron AC, Owen RG, Morgan GJ. Minimal residual

disease monitoring in multiple myeloma. Best Pract Res Clin Haematol

2002;15:197-222.

29. Martinelli G, Terragna C, Lemoli RM, Cavo M, Benni M, Motta MR, et al.

Clinical and molecular follow-up by amplification of the CDR-III IgH region in

multiple myeloma patients after autologous transplantation of hematopoietic

CD34+ stem cells. Haematologica 1999;84:397-404.

30. Owen RG, Goulden NJ, Oakhill A, Shiach C, Evans PA, Potter MN, et al.

Comparison of fluorescent consensus IgH PCR and allele-specific

oligonucleotide probing in the detection of minimal residual disease in childhood

ALL. Br J Haematol 1997;97:457-9.

31. Boyd SD, Gaeta BA, Jackson KJ, Fire AZ, Marshall EL, Merker JD, et al.

Individual variation in the germline Ig gene repertoire inferred from variable

region gene rearrangements. J Immunol 2010;184:6986-92.

32. Freeman JD, Warren RL, Webb JR, Nelson BH, Holt RA. Profiling the T-

cell receptor beta-chain repertoire by massively parallel sequencing. Genome

Res 2009;19:1817-24.

Research. on April 13, 2018. © 2015 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on March 9, 2015; DOI: 10.1158/1078-0432.CCR-14-2841

Page 23: Is This the Time to Introduce Minimal Residual Disease in Multiple ...

23

33. Robins H, Desmarais C, Matthis J, Livingston R, Andriesen J, Reijonen

H, et al. Ultra-sensitive detection of rare T cell clones. J Immunol Methods

2012;375:14-9.

34. Logan AC, Zhang B, Narasimhan B, Carlton V, Zheng J, Moorhead M, et

al. Minimal residual disease quantification using consensus primers and high-

throughput IGH sequencing predicts post-transplant relapse in chronic

lymphocytic leukemia. Leukemia 2013;27:1659-65.

35. Faham M, Zheng J, Moorhead M, Carlton VE, Stow P, Coustan-Smith E,

et al. Deep-sequencing approach for minimal residual disease detection in

acute lymphoblastic leukemia. Blood 2012;120:5173-80.

36. Huff CA, Matsui W. Multiple myeloma cancer stem cells. J Clin Oncol

2008;26:2895-900.

37. Thiago LS, Perez-Andres M, Balanzategui A, Sarasquete ME, Paiva B,

Jara-Acevedo M, et al. Circulating clonotypic B cells in multiple myeloma and

monoclonal gammopathy of undetermined significance. Haematologica

2014;99:155-62.

38. Ferrero S, Drandi D, Mantoan B, Ghione P, Omede P, Ladetto M.

Minimal residual disease detection in lymphoma and multiple myeloma: impact

on therapeutic paradigms. Hematol Oncol 2011;29:167-76.

39. Sarasquete ME, Garcia-Sanz R, Gonzalez D, Martinez J, Mateo G,

Martinez P, et al. Minimal residual disease monitoring in multiple myeloma: a

comparison between allelic-specific oligonucleotide real-time quantitative

polymerase chain reaction and flow cytometry. Haematologica 2005;90:1365-

72.

Research. on April 13, 2018. © 2015 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on March 9, 2015; DOI: 10.1158/1078-0432.CCR-14-2841

Page 24: Is This the Time to Introduce Minimal Residual Disease in Multiple ...

24

40. Corradini P, Cavo M, Lokhorst H, Martinelli G, Terragna C, Majolino I, et

al. Molecular remission after myeloablative allogeneic stem cell transplantation

predicts a better relapse-free survival in patients with multiple myeloma. Blood

2003;102:1927-9.

41. Korthals M, Sehnke N, Kronenwett R, Bruns I, Mau J, Zohren F, et al.

The level of minimal residual disease in the bone marrow of patients with

multiple myeloma before high-dose therapy and autologous blood stem cell

transplantation is an independent predictive parameter. Biol Blood Marrow

Transplant 2012;18:423-31 e3.

42. Langerak AW, Groenen PJ, Bruggemann M, Beldjord K, Bellan C,

Bonello L, et al. EuroClonality/BIOMED-2 guidelines for interpretation and

reporting of Ig/TCR clonality testing in suspected lymphoproliferations.

Leukemia 2012;26:2159-71.

43. van der Velden VH, Cazzaniga G, Schrauder A, Hancock J, Bader P,

Panzer-Grumayer ER, et al. Analysis of minimal residual disease by Ig/TCR

gene rearrangements: guidelines for interpretation of real-time quantitative PCR

data. Leukemia 2007;2:604-11.

44. Biran N, Ely S, Chari A. Controversies in the assessment of minimal

residual disease in multiple myeloma: clinical significance of minimal residual

disease negativity using highly sensitive techniques. Curr Hematol Malig Rep

2014;9:368-78.

45. Cooke F, Bakkus M, Thielemans K, Pico JL, Apperley JF, Samson D.

Use of quantitative ASO-PCR to predict relapse in multiple myeloma. Br J

Haematol 1999;105:317-9.

Research. on April 13, 2018. © 2015 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on March 9, 2015; DOI: 10.1158/1078-0432.CCR-14-2841

Page 25: Is This the Time to Introduce Minimal Residual Disease in Multiple ...

25

46. Garcia-Sanz R, Lopez-Perez R, Langerak AW, Gonzalez D, Chillon MC,

Balanzategui A, et al. Heteroduplex PCR analysis of rearranged

immunoglobulin genes for clonality assessment in multiple myeloma.

Haematologica 1999;84:328-35.

47. Kosmas C, Stamatopoulos K, Stavroyianni N, Zoi K, Belessi C, Viniou N,

et al. Origin and diversification of the clonogenic cell in multiple myeloma:

lessons from the immunoglobulin repertoire. Leukemia 2000;14:1718-26.

48. Sahota SS, Leo R, Hamblin TJ, Stevenson FK. Myeloma VL and VH

gene sequences reveal a complementary imprint of antigen selection in tumor

cells. Blood 1997;89:219-26.

49. Puig N, Sarasquete ME, Balanzategui A, Martinez J, Paiva B, Garcia H,

et al. Critical evaluation of ASO RQ-PCR for minimal residual disease

evaluation in multiple myeloma. A comparative analysis with flow cytometry.

Leukemia 2014;28:391-7.

50. Puig N, Sarasquete ME, Alcoceba M, Balanzategui A, Chillon MC,

Sebastian E, et al. The use of CD138 positively selected marrow samples

increases the applicability of minimal residual disease assessment by PCR in

patients with multiple myeloma. Ann Hematol 2013;92:97-100.

51. Martinez-Lopez J, Lahuerta JJ, Pepin F, Gonzalez M, Barrio S, Ayala R,

et al. Prognostic value of deep sequencing method for minimal residual disease

detection in multiple myeloma. Blood 2014;123:3073-9.

52. Boyd SD, Marshall EL, Merker JD, Maniar JM, Zhang LN, Sahaf B, et al.

Measurement and clinical monitoring of human lymphocyte clonality by

massively parallel VDJ pyrosequencing. Sci Transl Med 2009;1:12ra23.

Research. on April 13, 2018. © 2015 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on March 9, 2015; DOI: 10.1158/1078-0432.CCR-14-2841

Page 26: Is This the Time to Introduce Minimal Residual Disease in Multiple ...

26

53. Martinez-Lopez J, Fulciniti M, Barrio S, Carlton V, Moorhead M, Lahuerta

JJ, et al. Deep sequencing reveals oligoclonality at the immunoglobulin locus in

multiple myeloma patients [abstract]. In: Proceedings of the 55th ASH Annual

Meeting and Exposition; 2013 Dec 7-10; New Orleans, LA. Washington (DC):

American Society of Hematology. Abstract nr 401.

54. van Dongen JJ, Langerak AW, Bruggemann M, Evans PA, Hummel M,

Lavender FL, et al. Design and standardization of PCR primers and protocols

for detection of clonal immunoglobulin and T-cell receptor gene recombinations

in suspect lymphoproliferations: report of the BIOMED-2 Concerted Action

BMH4-CT98-3936. Leukemia 2003;17:2257-317.

55. San Miguel JF, Almeida J, Mateo G, Blade J, Lopez-Berges C, Caballero

D, et al. Immunophenotypic evaluation of the plasma cell compartment in

multiple myeloma: a tool for comparing the efficacy of different treatment

strategies and predicting outcome. Blood 2002;99:1853-6.

56. Paiva B, Vidriales MB, Cervero J, Mateo G, Perez JJ, Montalban MA, et

al. Multiparameter flow cytometric remission is the most relevant prognostic

factor for multiple myeloma patients who undergo autologous stem cell

transplantation. Blood 2008;112:4017-23.

57. Rawstron AC, Child JA, de Tute RM, Davies FE, Gregory WM, Bell SE,

et al. Minimal residual disease assessed by multiparameter flow cytometry in

multiple myeloma: impact on outcome in the Medical Research Council

Myeloma IX Study. J Clin Oncol 2013;31:2540-7.

58. Paiva B, Gutierrez NC, Rosinol L, Vidriales MB, Montalban MA, Martinez-

Lopez J, et al. High-risk cytogenetics and persistent minimal residual disease by

multiparameter flow cytometry predict unsustained complete response after

Research. on April 13, 2018. © 2015 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on March 9, 2015; DOI: 10.1158/1078-0432.CCR-14-2841

Page 27: Is This the Time to Introduce Minimal Residual Disease in Multiple ...

27

autologous stem cell transplantation in multiple myeloma. Blood 2012;119:687-

91.

59. Roussel M, Lauwers-Cances V, Robillard N, Hulin C, Leleu X,

Benboubker L, et al. Front-line transplantation program with lenalidomide,

bortezomib, and dexamethasone combination as induction and consolidation

followed by lenalidomide maintenance in patients with multiple myeloma: a

phase II study by the Intergroupe Francophone du Myelome. J Clin Oncol

2014;32:2712-7.

60. Corradini P, Voena C, Astolfi M, Ladetto M, Tarella C, Boccadoro M, et

al. High-dose sequential chemoradiotherapy in multiple myeloma: residual

tumor cells are detectable in bone marrow and peripheral blood cell harvests

and after autografting. Blood 1995;85:1596-602.

61. Bjorkstrand B, Ljungman P, Bird JM, Samson D, Gahrton G. Double

high-dose chemoradiotherapy with autologous stem cell transplantation can

induce molecular remissions in multiple myeloma. Bone Marrow Transplant

1995;15:367-71.

62. Swedin A, Lenhoff S, Olofsson T, Thuresson B, Westin J. Clinical utility

of immunoglobulin heavy chain gene rearrangement identification for tumour

cell detection in multiple myeloma. Br J Haematol 1998;103:1145-51.

63. Corradini P, Voena C, Tarella C, Astolfi M, Ladetto M, Palumbo A, et al.

Molecular and clinical remissions in multiple myeloma: role of autologous and

allogeneic transplantation of hematopoietic cells. J Clin Oncol 1999;17:208-15.

64. Martinelli G, Terragna C, Zamagni E, Ronconi S, Tosi P, Lemoli R, et al.

Polymerase chain reaction-based detection of minimal residual disease in

Research. on April 13, 2018. © 2015 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on March 9, 2015; DOI: 10.1158/1078-0432.CCR-14-2841

Page 28: Is This the Time to Introduce Minimal Residual Disease in Multiple ...

28

multiple myeloma patients receiving allogeneic stem cell transplantation.

Haematologica 2000;85:930-4.

65. Martinelli G, Terragna C, Zamagni E, Ronconi S, Tosi P, Lemoli RM, et

al. Molecular remission after allogeneic or autologous transplantation of

hematopoietic stem cells for multiple myeloma. J Clin Oncol 2000;18:2273-81.

66. Cavo M, Terragna C, Martinelli G, Ronconi S, Zamagni E, Tosi P, et al.

Molecular monitoring of minimal residual disease in patients in long-term

complete remission after allogeneic stem cell transplantation for multiple

myeloma. Blood 2000;96:355-7.

67. Ladetto M, Donovan JW, Harig S, Trojan A, Poor C, Schlossnan R, et al.

Real-Time polymerase chain reaction of immunoglobulin rearrangements for

quantitative evaluation of minimal residual disease in multiple myeloma. Biol

Blood Marrow Transplant 2000;6:241-53.

68. Bakkus MH, Bouko Y, Samson D, Apperley JF, Thielemans K, Van

Camp B, et al. Post-transplantation tumour load in bone marrow, as assessed

by quantitative ASO-PCR, is a prognostic parameter in multiple myeloma. Br J

Haematol 2004;126:665-74.

69. Martinez-Sanchez P, Montejano L, Sarasquete ME, Garcia-Sanz R,

Fernandez-Redondo E, Ayala R, et al. Evaluation of minimal residual disease in

multiple myeloma patients by fluorescent-polymerase chain reaction: the

prognostic impact of achieving molecular response. Br J Haematol

2008;142:766-74.

70. Ladetto M, Pagliano G, Ferrero S, Cavallo F, Drandi D, Santo L, et al.

Major tumor shrinking and persistent molecular remissions after consolidation

Research. on April 13, 2018. © 2015 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on March 9, 2015; DOI: 10.1158/1078-0432.CCR-14-2841

Page 29: Is This the Time to Introduce Minimal Residual Disease in Multiple ...

29

with bortezomib, thalidomide, and dexamethasone in patients with autografted

myeloma. J Clin Oncol 2010;28:2077-84.

71. Putkonen M, Kairisto V, Juvonen V, Pelliniemi TT, Rauhala A, Itala-

Remes M, et al. Depth of response assessed by quantitative ASO-PCR predicts

the outcome after stem cell transplantation in multiple myeloma. Eur J Haematol

2010;85:416-23.

72. Ferrero S, Ladetto M, Drandi D, Cavallo F, Genuardi E, Urbano M, et al.

Long-term results of the GIMEMA VEL-03-096 trial in MM patients receiving

VTD consolidation after ASCT: MRD kinetics' impact on survival. Leukemia

2014 Jul 6. [Epub ahead of print].

73. Lipinski E, Cremer FW, Ho AD, Goldschmidt H, Moos M. Molecular

monitoring of the tumor load predicts progressive disease in patients with

multiple myeloma after high-dose therapy with autologous peripheral blood

stem cell transplantation. Bone Marrow Transplant 2001;28:957-62.

74. Lioznov M, Badbaran A, Fehse B, Bacher U, Zander AR, Kroger NM.

Monitoring of minimal residual disease in multiple myeloma after allo-SCT: flow

cytometry vs PCR-based techniques. Bone Marrow Transplant 2008;41:913-6.

75. Ladetto M, Bruggemann M, Monitillo L, Ferrero S, Pepin F, Drandi D, et

al. Next-generation sequencing and real-time quantitative PCR for minimal

residual disease detection in B-cell disorders. Leukemia 2014;28:1299-307.

76. Galimberti S, Benedetti E, Morabito F, Papineschi F, Callea V, Fazzi R,

et al. Prognostic role of minimal residual disease in multiple myeloma patients

after non-myeloablative allogeneic transplantation. Leuk Res 2005;29:961-6.

Research. on April 13, 2018. © 2015 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on March 9, 2015; DOI: 10.1158/1078-0432.CCR-14-2841

Page 30: Is This the Time to Introduce Minimal Residual Disease in Multiple ...

30

Table 1. Summary of the most relevant studies based on MFC detection of MRD in MM. It should be noted that albeit most of the published

results were obtained by former (4-7 color) and less sensitive (10-4) MFC approaches, the persistence of MRD is consistently associated with a

significantly shorter PFS and often OS as compared to MRD-negative patients.

REFERENCE N SETTING METHOD LOD APPLICAB IMMUNOPHENOTYPIC REMISSION

PFS ACCORDING

TO MRD P

OS ACCORDING

TO MRD P

San Miguel et al., 2002 (22) 87 CT or CT+ASCT

4-color MFC 10-4 NA 26% 60m vs 34m 0.02 NA -

Rawstron et al., 2002 (20) 45 ASCT 3-color MFC 10-3 - 10-4 94% 56% (25/45) 35m vs 20m 0.03 76% vs 64% at

5-years 0.28

Paiva et al., 2008 (56) 295 CT+ASCT 4-color MFC 10-4 ~95% 42% (125/295) 71m vs 37m <0.001 NR vs 89m 0.002

Paiva et al., 2011 (7) 102 VMP or VTP 4-color MFC 10-4 - 10-5 ~95% 43% (44/102) 90% vs 35% at

3-years <0.001 94% vs 70% at 3-years 0.08

Paiva et al., 2012 (58) 241 (CR)

CTn or TD or CT/Btz or

VTD +ASCT

4-color MFC 10-4 - 10-5 ~95% 74% (154/241) 86% vs 58% at

3-years <0.001 94% vs 80% at 3-years 0.001

Rawston et al., 2013 (57) 397 CTD or CVAD + ASCT

6-color MFC 10-4 NA 62% (247/397) 29m vs 16m <0.001 81m vs 59m 0.02

Roussel et al., 2014 (59) 31 VRD + ASCT + VRD + Len

7-color MFC 10-5 NA 68% (21/31) 100% vs 30% at

3-years NA NA -

MFC: multiparameter flow cytometry; LOD: limit of detection; NA: not available; PFS: progression free survival; MRD: minimal residual disease;

OS: overall survival; ASCT: autologous stem cell transplantation; CT: VBMCP, VBAD; VMP: bortezomib, melphalan, prednisone; VTP:

bortezomib, thalidomide, prednisone; TD: thalidomide, dexamethasone; VTD: bortezomib, thalidomide, dexamethasone; CTD:

cyclophosphamide, thalidomide, dexamethasone; CVAD: cyclophosphamide, vincristine, doxorubicin, dexamethasone; VRD: bortezomib,

lenalidomide, dexamethasone; Len: lenalidomide; CR: complete response.

Research.

on April 13, 2018. ©

2015 Am

erican Association for C

ancerclincancerres.aacrjournals.org

Dow

nloaded from

Author m

anuscripts have been peer reviewed and accepted for publication but have not yet been edited.

Author M

anuscript Published O

nlineFirst on M

arch 9, 2015; DO

I: 10.1158/1078-0432.CC

R-14-2841

Page 31: Is This the Time to Introduce Minimal Residual Disease in Multiple ...

31

Table 2. Summary of the most relevant studies based on PCR detection of MRD in MM. The low number of cases in most of the series is

probably due to the technical problems related to this technique. However, in all studies the persistence of MRD was associated with a

significantly shorter PFS and often OS as compared to MRD-negative patients.

REFERENCE N SETTING METHOD LOD APPLICAB MOLECULAR REMISSION

PFS ACCORDING

TO MRD P

OS ACCORDING

TO MRD P

Martinelli et al., 2000 (65) 50 ASCT o ALLO ASO 10-5 88% (44/50) 27% (12/44) 110m vs 35m <.005 NA -

Ladetto et al., 2000 (67) 29 ASCT RT NESTED

10-4 10-3

66% 73-100% NA NA - NA -

Corradini et al., 2003 (40) 70 ALLO ASO 10-6 69% 33% (16/48) 100% vs 0%1 NA -

Bakkus et al., 2004 (68) 87 ASCT ASO 10-4 77% 35% (21/60)≤ 0.015% 65% (39/60) >0.015% 64m vs 16m 0.001 NA ns

Galimberti et al., 2005 (76) 20 ASCT+ALLO fASO 10-3-10-5 NA 15% (3)

60% (12) NA - 76% vs 34% at 2ys .03

Sarasquete et al., 2005 (39) 24 ASCT ASO 5x10-5-10-

5 75% (24/32) 29% (7/24) 34m vs 15m .042 NA -

Martínez-Sánchez et al., 2008 (69) 53 ASCT fASO 10-3-10-4 91% 53% (28/53) 68% vs 28% .001 86% vs 68% ns

Putkonen et al., 2010 (71) 37 ASCT&ALLO RQ 10-4-10-5 86% 53% (16/30)

71% (5/7) 70m vs 19m .003 median not reached .1

Ladetto, et al., 2010 (70) Ferrero et al., 2014 (72) 39 ASCT+VTD RQ

NESTED 10-6 51% 18% NR vs 38mo vs 9 ms2 <.001 72% vs 48% at

8 ys .041

Korthals et al., 2012 (41) 53 ASCT RQ-ASO 10-4-10-5 78% 48% (26/53) 35 vs 20 .001 70 vs 45 .04

Puig et al., 2014 (49) 103 ASCT RQ-ASO 10-5 42% 46% NR vs 31mo .002 NR vs 60mo3 .008

PCR: polymerase chain reaction; NA: not available; fASO: fluorescent PCR; LOD: limit of detection; APPLICAB: applicability; PFS: progression

free survival; MRD: minimal residual disease; OS: overall survival; ASCT: autologous stem cell transplantation; ALLO: allogeneic stem cell

Research.

on April 13, 2018. ©

2015 Am

erican Association for C

ancerclincancerres.aacrjournals.org

Dow

nloaded from

Author m

anuscripts have been peer reviewed and accepted for publication but have not yet been edited.

Author M

anuscript Published O

nlineFirst on M

arch 9, 2015; DO

I: 10.1158/1078-0432.CC

R-14-2841

Page 32: Is This the Time to Introduce Minimal Residual Disease in Multiple ...

32

transplantation; ASO: allelic specific oligonucleotide PCR; m: months; ns: non significant; VTD: bortezomib (V), thalidomide(T) y

dexamethasone (D); RQ: real-time quantitative PCR; The cumulative risk of relapse was 0% for PCR-negative patients and 100% for PCR-

positive patients; median remission duration was not reached for patients in major MRD response, 38 months for those experiencing MRD

reappearance and 9 months for patients with MRD persistence (P<0.001); among patients in CR.

Research.

on April 13, 2018. ©

2015 Am

erican Association for C

ancerclincancerres.aacrjournals.org

Dow

nloaded from

Author m

anuscripts have been peer reviewed and accepted for publication but have not yet been edited.

Author M

anuscript Published O

nlineFirst on M

arch 9, 2015; DO

I: 10.1158/1078-0432.CC

R-14-2841

Page 33: Is This the Time to Introduce Minimal Residual Disease in Multiple ...

Published OnlineFirst March 9, 2015.Clin Cancer Res   Bruno Paiva, Noemi Puig, Ramón García-Sanz, et al.   Multiple Myeloma Clinical Practice?Is This the Time to Introduce Minimal Residual Disease in

  Updated version

  10.1158/1078-0432.CCR-14-2841doi:

Access the most recent version of this article at:

  Manuscript

Authoredited. Author manuscripts have been peer reviewed and accepted for publication but have not yet been

   

   

   

  E-mail alerts related to this article or journal.Sign up to receive free email-alerts

  Subscriptions

Reprints and

  [email protected] at

To order reprints of this article or to subscribe to the journal, contact the AACR Publications

  Permissions

  Rightslink site. Click on "Request Permissions" which will take you to the Copyright Clearance Center's (CCC)

.http://clincancerres.aacrjournals.org/content/early/2015/03/07/1078-0432.CCR-14-2841To request permission to re-use all or part of this article, use this link

Research. on April 13, 2018. © 2015 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on March 9, 2015; DOI: 10.1158/1078-0432.CCR-14-2841