Ultra high-risk myeloma can be defined as myeloma leading to death within 24 months. Despite tremendous improvements in the past decade (especially because of the availability of novel drugs such as thalidomide, bortezomib, and lenalidomide), these patients still represent 15% to 20% of the patients. Many prognostic factors can help to define these patients, including age, renal insufficiency, poor performance status, comorbities, International Staging System (ISS) stage 3, high proliferation, leukemic presentation, and acquired genetic changes, as defined by interphase fluorescence in situ hybridization or genomics. Several combinations of these prognostic parameters can define ultra high-risk patients, making a universal therapeutic proposal almost impossible. However, focusing on fit patients with ISS 3, high proliferation, and poor-risk genetic changes, these patients should probably benefit from dose-dense and prolonged therapeutic schemas, ideally within prospective trials.

For a long time, the prognosis of multiple myeloma (MM) has been poor with limited treatment options. Many studies reported a median overall survival of less than 3 years. The first glimpse of improved outcomes was reports in the 1990s of the use of high-dose melphalan with stem cell support.1–3  Using a polychemotherapy-based induction, followed by one or two courses of high-dose melphalan, the median overall survival improved to 5 years. However, more than one-half of the patients are not eligible for these intensive approaches. In the past decade, tremendous progresses have been observed, with the availability of three major new drugs: (1) thalidomide, (2) bortezomib, and (3) lenalidomide. With the use of these drugs, most commonly in combination with high-dose melphalan, patients now display a much better outcome.4–10  The latest estimates of median overall survival are probably around 10 years for patients under age 65 and around 5 to 6 years for older patients (Intergroupe Francophone du Myélome [IFM], personal communication, 2010). However, despite this remarkable progress, we have recognized that not all patients enjoy such a long survival. Some patients present with highly refractory MM or exhibit early relapses after an initial response. The three objectives of this paper are to (1) try and define who these patients are, (2) recognize these patients at the time of diagnosis, and (3) propose specific therapeutic approaches to improve their outcome.

The definition of these ultra high-risk patients is by definition arbitrary. This review focuses on patients with a median overall survival of less than 2 years. Many papers have described a huge number of prognostic factors in MM. Among this list, we can focus on those who have been confirmed by several studies. A nonexhaustive list comprises a high serum β2-microglobulin; a high C-reactive protein level; a low albumin level; a high creatinine level; a high lactate dehydrogenase level; a low hemoglobin level; a low platelet count; a high labeling index; leukemic presentation; poor cytogenetic parameters (eg, t(4;14), del(17p), or t(14;16)); and, as usual in oncology, older age. In this list, the most important parameters are probably β2-microglobulin,11–13  proliferation,14–16  and genetic abnormalities (Table 1).17–21  In a large cohort of more than 12,000 patients, the International Myeloma Working Group described an International Staging System (ISS) based on β2-microglobulin and albumin levels.13  This prognostic model classifies the patients into three groups, with different overall survival (62 months, 44 months, and 29 months for stages 1, 2, and 3, respectively). This system allows great progress in prognostication at the population level, but is more limited at the individual level. The weakness of this model is that it does not take into account intrinsic prognostic parameters, such as proliferation or genetic abnormalities. An ideal prognostic model would probably combine β2-microglobulin level (or ISS) that reflects tumor mass, renal insufficiency, and general patient condition, a marker of plasma cell proliferation (measured by labeling index or conventional cytogenetics) and genetic changes. The best way to identify these genetic changes is so far not defined. Many techniques can be used, including interphase fluorescence in situ hybridization (iFISH),17–21  gene expression profiling (GEP),22–24  or DNA copy number changes (analyzed by comparative genome hybridization [CGH] array or single nucleotide polymorphism [SNP] array).25,26  Most large series have used iFISH. The weakness of this technique in MM is that it requires plasma cell identification and that it allows the analysis of only a small number of chromosomal abnormalities. Despite these limitations, it has been shown that t(4;14) and del(17p) are major prognostic factors, at least in patients treated before the novel drug era.17–21  An updated ISS effort did show that karyotype abnormalities (reflecting high proliferation) and genetic changes (as identified by iFISH) significantly improved the ISS model (Enhancement of ISS Staging System incorporating genetic changes: an International Myeloma Working Group (IMWG) collaborative project, unpublished data). High-throughput molecular technologies (GEP and CGH array/SNP array) are probably more powerful, because they analyze all the abnormalities observed throughout the whole genome. However, these technologies require plasma cell purification and highly specifically dedicated platforms (including bioinformatics), and are not widely available to most physicians.

Table 1.

Summary of the most prognostic parameters defining ultra high-risk patients

Summary of the most prognostic parameters defining ultra high-risk patients
Summary of the most prognostic parameters defining ultra high-risk patients

The next question is the stability of these prognostic factors in the novel drug era. Most (if not all) of them have been defined in retrospective cohorts of patients treated before the wide use of thalidomide, bortezomib, and lenalidomide. The current question is whether these prognostic parameters are still valid with novel treatment modalities?

The ISS is probably still prognostic with novel combinations. Even though few studies specifically addressed this issue, most of the recently published trials showed that patients with ISS stage 3 displayed a shorter survival.7,10,27  This statement is probably also true for highly proliferative disease. Here, again, few studies did analyze this question. In patients treated at the University of Arkansas Total Therapy 3 program (polychemotherapy induction, double intensive melphalan, consolidation, and long-term maintenance with bortezomib-lenalidomide-dexamethasone cycles), an abnormal karyotype (indirectly reflecting plasma cell proliferation) is a powerful prognostic parameter.27  Similarly, a small study from the Mayo Clinic suggested that a high labeling index was still prognostic in patients treated with lenalidomide-dexamethasone.16 

The same question addressed to genetic changes is still evolving. Some preliminary reports suggested that bortezomib was able to overcome the prognostic value of t(4;14). This has been first suggested in the VISTA (Velcade as Initial Standard Therapy in Multiple Myeloma) trial (comparing melphalan-prednisone vs bortezomib-melphalan-prednisone).28  In a very small number of patients (26 patients) presenting either t(4;14), del(17p), or t(14;16), time to progression and overall survival were similar in the bortezomib arm to that of patients lacking these chromosomal abnormalities (limiting the stability of the conclusions given limited power). The second report came from the Arkansas Total Therapy 3 program that showed that t(4;14), but also del(17p), were not anymore prognostic factors with this very peculiar therapy program.27,29  More recently, the IFM did show that a short-term bortezomib-dexamethasone (BD) induction significantly improved the outcome (both for event-free survival and overall survival) of patients with t(4;14), but not that of patients with del(17p).30  Nevertheless, even among bortezomib-treated patients, the t(4;14)(p16;q32) remains a significant prognostic factor Thus, based on iFISH and GEP analyses, t(4;14) is probably no longer a (strong) prognostic factor in patients treated with bortezomib. Because the VISTA trial and the Arkansas results showed a complete overcoming of the prognostic value of t(4;14), in contrast to the IFM data that showed only a partial improvement, it can be suggested that long-term treatment with bortezomib is recommended for these patients. Further validation, however, is needed. The case of patients with del(17p) is less clear. All of the recently published reports showed that del(17p) remains a strong prognostic factor.16,31  The only study suggesting that del(17p) was no longer prognostic came from the Arkansas group.29  Based on GEP analysis, using a surrogate proposal to detect -17p13, they showed that patients with a low TP53 expression encountered a similar outcome than patients with normal TP53 expression in the Total Therapy 3 program.

Some authors (especially those from Arkansas University) claim that all the patients should be analyzed by GEP to define a really strong prediction for survival. The first question is the feasibility of GEP in the routine practice. In a multicenter setting, only about half of the patients are analyzable with this technique (Avet-Loiseau H, Institut de Biologie, oral communication, September 17, 2010), because of the quality of the bone marrow samples sent to the laboratory. The second question is the definition of ultra high-risk patients using these technologies. Two reports on large cohorts of patients analyzed with GEP have been published so far (Figure 1).22,24  These two studies defined two different sets of genes identifying high-risk patients. Whether these models are identifying the same group of high-risk patients is an unresolved question. First, the two models were built on different platforms. Moreover, these models have been built on patients treated very differently. The Arkansas model was based on patients treated in the Total Therapy 2 program (polychemotherapy induction, double intensive melphalan, consolidation, long-term thalidomide maintenance), whereas the IFM model was built on patients treated with a vincristine-adriamycin-dexamethasone (VAD) induction, followed by double high-dose melphalan. A temptative cross-validation has been performed and showed a weak confirmation. This can be due to the different platforms, but also to the different therapeutic strategies. Despite these technical limitations, it seems that GEP is promising to define high-risk patients. Another study from the IFM using SNP array did show that some genomic changes (eg, 1q gains, trisomy 5, or 12p deletions) allowed identification of patients with very different outcomes.26  But, again, this technology requires plasma cell purification and is probably not usable for all the patients in a multicenter setting.

Figure 1.

Genomic data showing the predictability of ultra high-risk myeloma patients using genomics. (A) Arkansas model using gene expression profiling. (B) IFM model using the SNP array.

Figure 1.

Genomic data showing the predictability of ultra high-risk myeloma patients using genomics. (A) Arkansas model using gene expression profiling. (B) IFM model using the SNP array.

Close modal

Ultra high-risk patients could be defined by ISS stage 3, a high plasma cell proliferation, and the presence of specific genetic changes, including del(17p) and probably other genomic abnormalities that need further work for a widely accepted definition. A specific statement is mandatory for the rare patients presenting with primary plasma cell leukemia. Even though no large study specifically dedicated for this patient population exists in the literature, most authors agree to consider these patients as high-risk (and probably ultra high-risk).32–34  Actually, these patients often combine a high β2-microglobulin level (or ISS stage 3), a high proliferative index reflected by the frequent abnormal karyotype and poor risk genetic abnormalities.

Even though the definition of ultra high-risk MMs varies from institution to institution, their management is very disappointing. For instance, in the Arkansas definition based on GEP, these patients represent 13% of all MMs. Even using extremely intensive and prolonged therapy, these patients display a very poor outcome.22  Using more “classical” therapies based on bortezomib-dexamethasone induction, followed by one or two courses of high-dose melphalan for younger patients, patients with del(17p) present a short, event-free survival and overall survival. Thus, how can this poor outcome be improved?

Several strategies have been tested. If considering the youngest patients (younger than age 65 or 70), several induction combinations (before high-dose therapy) have addressed this issue. These combinations included VAD, TD (thalidomide-dexamethasone), BD, or more complex strategies such as VTD-PACE (bortezomib-thalidomide-dexamethasone-cisplatinum-adriamycin-cyclophosphamide-etoposide). Despite some improvements in the complete response rates, a similar number of patients still died rapidly in all combination schemes. Then, some investigators worked on the high-dose regimen. A double-intensive strategy does not seem to benefit these patients.3  A combination of high-dose melphalan with bortezomib seemed to increase the response rate, but no data are available for overall survival.35  Finally, different consolidation/maintenance strategies have been tested. Thalidomide maintenance is definitely not the solution.36,37  Long-term maintenance with lenalidomide seems to improve the progression-free survival, but no data are available for overall survival (that could be shortened by the appearance of specific or overall resistances at relapse) or for high-risk patients (Attal M, personal communication, August 2010). Iterative long-term reinductions with bortezomib-lenalidomide-dexamethasone combinations appear promising for patients with t(4;14) or del(17p), but a population (representing about 13% of the patients) still escapes to this strategy and presents early death (Table 2).38 

Table 2.

Suggestions for treatment of ultra high-risk patients

Suggestions for treatment of ultra high-risk patients
Suggestions for treatment of ultra high-risk patients

If a solution does exist, it will definitely be found through specific therapeutic trials. For instance, a trial testing dose-dense chemotherapy, prolonged over a long period of time, could be a solution to try to maintain a continuous therapeutic pressure on the clone. Another possibility could be to propose a frontline myeloablative stem cell transplant for such patients younger than age 50. These phase 2 trials should typically be multicentric, possibly international, to test different approaches over a short period of time. An important limit of this “trial” approach is that many of these poor-risk patients are not eligible for trial enrollment because of poor performance status, renal insufficiency, or comorbidities. Some investigators propose to test new drugs in these ultra high-risk patients. This proposal is highly debatable. The risk is to kill promising drugs tested on highly resistant patients. A strategy is recommended in which modern prognostic parameters (including genomics) would be analyzed in phase 2 and phase 3 trials testing novel drugs—eg, carfilzomib (second-generation proteasome inhibitor), pomalidomide (novel IMID [imidazoline]), heat shock protein, or histone deacetylase inhibitors—to specifically analyze their efficacy on high-risk patients.

Conflict-of-interest disclosure: The author is on the Board of Directors and Advisory Committees for Janssen-Cilag and Celgene.

Off-label drug use: None disclosed.

Hervé Avet-Loiseau MD, PhD, Professor, Laboratoire d'Hématologie, Institut de Biologie, 9 Quai Moncousu, 44093 Nantes Cedex 09, France; Phone: +33240087774; Fax: +33240084050; e-mail: herve.avetloiseau@chu-nantes.fr

1
Attal
M
Harousseau
JL
Stoppa
AM
et al
A prospective, randomized trial of autologous bone marrow transplantation and chemotherapy in multiple myeloma. Intergroupe Français du Myélome
N Engl J Med
1996
335
91
97
2
Child
JA
Morgan
GJ
Davies
FE
et al
High-dose chemotherapy with hematopoietic stem-cell rescue for multiple myeloma
N Engl J Med
2003
348
1875
1883
3
Attal
M
Harousseau
JL
Facon
T
et al
Single versus double autologous stem-cell transplantation for multiple myeloma
N Engl J Med
2003
349
2495
2502
4
Palumbo
A
Bringhen
S
Caravita
T
et al
Oral melphalan and prednisone chemotherapy plus thalidomide compared with melphalan and prednisone alone in elderly patients with multiple myeloma: randomised controlled trial
Lancet
2006
367
825
831
5
Facon
T
Mary
JY
Hulin
C
et al
Melphalan and prednisone plus thalidomide versus melphalan and prednisone alone or reduced-intensity autologous stem cell transplantation in elderly patients with multiple myeloma (IFM 99–06): a randomised trial
Lancet
2007
370
1209
1218
6
Richardson
PG
Sonneveld
P
Schuster
MW
et al
Bortezomib or high-dose dexamethasone for relapsed multiple myeloma
N Engl J Med
2005
352
2487
2498
7
Harousseau
JL
Attal
M
Avet-Loiseau
H
et al
Bortezomib plus dexamethasone is superior to vincristine plus doxorubicin plus dexamethasone as induction treatment prior to autologous stem-cell transplantation in newly diagnosed multiple myeloma: results of the IFM 2005-01 Phase III Trial
J Clin Oncol
2010
9
7
[Epub ahead of print]
8
Dimopoulos
M
Spencer
A
Attal
M
et al
Lenalidomide plus dexamethasone for relapsed or refractory multiple myeloma
N Engl J Med
2007
357
2123
2132
9
Weber
DM
Chen
C
Niesvizky
R
et al
Lenalidomide plus dexamethasone for relapsed multiple myeloma in North America
N Engl J Med
2007
357
2133
2142
10
Rajkumar
SV
Jacobus
S
Callander
NS
et al
Lenalidomide plus high-dose dexamethasone versus lenalidomide plus low-dose dexamethasone as initial therapy for newly diagnosed multiple myeloma: an open-label randomised controlled trial
Lancet Oncol
2010
11
29
37
11
Bataille
R
Durie
BG
Grenier
J
Serum beta2 microglobulin and survival duration in multiple myeloma: a simple reliable marker for staging
Br J Haematol
1983
55
439
447
12
Durie
BG
Stock-Novack
D
Salmon
SE
et al
Prognostic value of pretreatment serum beta 2 microglobulin in myeloma: a Southwest Oncology Group Study
Blood
1990
75
823
830
13
Greipp
PR
San Miguel
J
Durie
BG
et al
International staging system for multiple myeloma
J Clin Oncol
2005
23
3412
3420
14
Greipp
PR
Katzmann
JA
O'Fallon
WM
Kyle
RA
Value of beta 2-microglobulin level and plasma cell labeling indices as prognostic factors in patients with newly diagnosed myeloma
Blood
1988
72
219
223
15
Shaughnessy
J
Jacobson
J
Sawyer
J
et al
Continuous absence of metaphase-defined cytogenetic abnormalities, especially of chromosome 13 and hypodiploidy, ensures long-term survival in multiple myeloma treated with Total Therapy I: interpretation in the context of global gene expression
Blood
2003
101
3849
3856
16
Kapoor
P
Kumar
S
Fonseca
R
et al
Impact of risk stratification on outcome among patients with multiple myeloma receiving initial therapy with lenalidomide and dexamethasone
Blood
2009
114
518
521
17
Facon
T
Avet-Loiseau
H
Guillerm
G
et al
Chromosome 13 abnormalities identified by FISH analysis and serum beta2-microglobulin produce a powerful myeloma staging system for patients receiving high-dose therapy
Blood
2001
97
1566
1571
18
Fonseca
R
Blood
E
Rue
M
et al
Clinical and biologic implications of recurrent genomic aberrations in myeloma
Blood
2003
101
4569
4575
19
Gertz
MA
Lacy
MQ
Dispenzieri
A
et al
Clinical implications of t(11;14)(q13;q32), t(4;14)(p16.3;q32), and -17p13 in myeloma patients treated with high-dose therapy
Blood
2005
106
2837
2840
20
Fonseca
R
Blood
E
Rue
M
et al
Clinical and biologic implications of recurrent genomic aberrations in myeloma
Blood
2003
101
4569
4575
21
Avet-Loiseau
H
Attal
M
Moreau
P
et al
Genetic abnormalities and survival in multiple myeloma: the experience of the Intergroupe Francophone du Myélome
Blood
2007
10
3489
3495
22
Shaughnessy
JD
Jr
Zhan
F
Burington
BE
et al
A validated gene expression model of high-risk multiple myeloma is defined by deregulated expression of genes mapping to chromosome 1
Blood
2007
109
2276
2284
23
Mulligan
G
Mitsiades
C
Bryant
B
et al
Gene expression profiling and correlation with outcome in clinical trials of the proteasome inhibitor bortezomib
Blood
2007
109
3177
3188
24
Decaux
O
Lodé
L
Magrangeas
F
et al
Prediction of survival in multiple myeloma based on gene expression profiles reveals cell cycle and chromosomal instability signatures in high-risk patients and hyperdiploid signatures in low-risk patients: a study of the Intergroupe Francophone du Myélome
J Clin Oncol
2008
26
4798
4805
25
Carrasco
DR
Tonon
G
Huang
Y
et al
High-resolution genomic profiles define distinct clinico-pathogenetic subgroups of multiple myeloma patients
Cancer Cell
2006
9
313
325
26
Avet-Loiseau
H
Li
C
Magrangeas
F
et al
Prognostic significance of copy-number alterations in multiple myeloma
J Clin Oncol
2009
27
4585
4590
27
Barlogie
B
Anaissie
E
van Rhee
F
et al
Incorporating bortezomib into upfront treatment for multiple myeloma: early results of total therapy 3
Br J Haematol
2007
138
176
185
28
San Miguel
JF
Schlag
R
Khuageva
NK
et al
Bortezomib plus melphalan and prednisone for initial treatment of multiple myeloma
N Engl J Med
2008
359
906
917
29
Shaughnessy
JD
Zhou
Y
Haessler
J
et al
TP53 deletion is not an adverse feature in multiple myeloma treated with Total Therapy 3
Br J Haematol
2009
147
347
351
30
Avet-Loiseau
H
Leleu
X
Roussel
M
et al
Bortezomib plus dexamethasone induction improves outcome of patients with t(4;14) myeloma but not outcome of patients with del(17p)
J Clin Oncol
2010
8
9
[Epub ahead of print]
31
Reece
D
Song
KW
Fu
T
et al
Influence of cytogenetics in patients with relapsed or refractory multiple myeloma treated with lenalidomide plus dexamethasone: adverse effect of deletion 17p13
Blood
2009
114
522
525
32
Avet-Loiseau
H
Daviet
A
Brigaudeau
C
et al
Cytogenetic, interphase, and multicolor fluorescence in situ hybridization analyses in primary plasma cell leukemia: a study of 40 patients at diagnosis, on behalf of the Intergroupe Francophone du Myélome and the Groupe Français de Cytogénétique Hématologique
Blood
2001
97
822
825
33
Musto
P
Rossini
F
Gay
F
et al
Efficacy and safety of bortezomib in patients with plasma cell leukemia
Cancer
2007
109
2285
2290
34
Ramsingh
G
Mehan
P
Luo
J
Vij
R
Morgensztern
D
Primary plasma cell leukemia: a Surveillance, Epidemiology, and End Results database analysis between 1973 and 2004
Cancer
2009
115
5734
5739
35
Roussel
M
Moreau
P
Huynh
A
et al
Bortezomib and high-dose melphalan as conditioning regimen before autologous stem cell transplantation in patients with de novo multiple myeloma: a phase 2 study of the Intergroupe Francophone du Myelome (IFM)
Blood
2010
115
32
37
36
Attal
M
Harousseau
JL
Leyvraz
S
et al
Maintenance therapy with thalidomide improves survival in patients with multiple myeloma
Blood
2006
108
3289
3294
37
Spencer
A
Prince
HM
Roberts
AW
et al
Consolidation therapy with low-dose thalidomide and prednisolone prolongs the survival of multiple myeloma patients undergoing a single autologous stem-cell transplantation procedure
J Clin Oncol
2009
27
1788
1793
38
Nair
B
van Rhee
F
Shaughnessy
JD
Jr
et al
Superior results of Total Therapy 3 (2003-33) in gene expression profiling-defined low-risk multiple myeloma confirmed in subsequent trial 2006-66 with bortezomib, lenalidomide and dexamethasone (VRD) maintenance
Blood
2010
2
2
[Epub ahead of print]