Patients with multiple myeloma have experienced a great improvement in survival over the past century because of the introduction of novel therapeutic strategies. However, a subgroup of patients with poorer outcomes than expected is considered high risk and identified by the presence of patient- and disease-based factors such as frailty, extramedullary disease, cytogenetic abnormalities, or even relapses occurring earlier than expected according to the baseline factors. Although the management of patients with high-risk features is not well established because of the lack of specific trials in this subgroup of patients and because of their underrepresentation in the clinical trials, treatment should be planned on 2 pillars: (1) poor prognosis with the presence of high-risk features can be at least improved or even abrogated by achieving a deep and sustained response over time, and (2) this can most likely be obtained through using the best therapeutic options and in a response-adapted way. Some clinical trials that have been planned or are ongoing include only patients with high-risk features, using the most effective therapies (proteasome inhibitors, immunomodulatory drugs, and anti-CD38 monoclonal antibodies) as well as chimeric antigen receptor T cells and T-cell engagers that will unravel what the best therapeutic approach will be to overcome the poor prognosis of the presence of high-risk features.

Learning Objectives

  • Identify high-risk myeloma based on patient-, disease-, or outcome-based factors

  • Be able to define the key objectives to overcome poor prognosis with the presence of high-risk features

  • Define the best therapeutic strategy for patients with high-risk features

A 48-year-old man with newly diagnosed (ND), Revised International Staging System (R-ISS) III (ISS III plus del(17p)), Bence-Jones κ multiple myeloma (MM) sought treatment and consultation for MM that had been diagnosed in another institution. The patient had an active lifestyle, and the workup showed mild anemia and small lytic lesions in the pelvis and femora as myeloma-defining events. His Eastern Cooperative Oncology Group performance status was 1. He was treated with 6 induction cycles of lenalidomide, bortezomib, and dexamethasone (RVd), achieving a very good partial response, followed by high-dose melphalan and autologous stem cell transplantation (HDM-ASCT), achieving stringent complete remission (sCR) with minimal residual disease (MRD) positivity. He rejected a second ASCT and proceeded to consolidation with 2 cycles of lenalidomide, carfilzomib, and dexamethasone, achieving sCR and MRD negativity. Maintenance with lenalidomide was prescribed. Twelve months after starting maintenance therapy, relapse occurred with reappearance of the M-component in urine. He was included in a clinical trial and treated with B-cell maturation antigen (BCMA) chimeric antigen receptor T (CAR-T) cells, and a new sCR and MRD negativity were achieved. The patient continues in follow-up.

Table 1 summarizes the most relevant patient- and disease-based factors to define high-risk patients.

Table 1.

Patient- and disease-based factors for the identification of high-risk MM

High-risk featuresDefinition
Patient-based factors  
 Frailty status IMWG frailty score
Modified IMWG frailty score
R-MCI
GAH 
Disease-based factors  
 Aggressiveness in the clinical presentation Extramedullary disease (no bone-related plasmacytomas)
Plasma cell leukemia
LDH elevated 
 Cytogenetic abnormalities del(17p), t(4;14), t(14;16), amp1q, del(1p) 
 Mutations TP53 
 Biochemical abnormalities LDH elevated
β2-microglobulin ≥5.5 mg/L
Albumin levels ≤3.5 mg/L 
Prognostic scores  
 R-ISS R-ISS III: beta2-microglobulin ≥5.5 mg/L plus either LDH elevated or high-risk CA (del(17p), t(4;14), or t(14;16)) 
High-risk featuresDefinition
Patient-based factors  
 Frailty status IMWG frailty score
Modified IMWG frailty score
R-MCI
GAH 
Disease-based factors  
 Aggressiveness in the clinical presentation Extramedullary disease (no bone-related plasmacytomas)
Plasma cell leukemia
LDH elevated 
 Cytogenetic abnormalities del(17p), t(4;14), t(14;16), amp1q, del(1p) 
 Mutations TP53 
 Biochemical abnormalities LDH elevated
β2-microglobulin ≥5.5 mg/L
Albumin levels ≤3.5 mg/L 
Prognostic scores  
 R-ISS R-ISS III: beta2-microglobulin ≥5.5 mg/L plus either LDH elevated or high-risk CA (del(17p), t(4;14), or t(14;16)) 

GAH, geriatric assessment in hematology; R-MCI, Revised Myeloma Comorbidity Index.

Frailty

For a long time, chronological age influenced treatment decisions, and the outcome was poor for the elderly. The International Myeloma Working Group (IMWG), through a pooled analysis including 869 ND elderly patients enrolled in clinical trials, built a simplified geriatric score based on age, comorbidities, and cognitive and physical conditions to distinguish among fit (score  =  0), intermediate fitness (score  =  1), and frailty (score ≥ 2). Frail patients showed a significantly shorter overall survival (OS; 57% at 3 years) than unfit (76% at 3 years) and fit patients (84% at 3 years).1  Many clinical trials are using an IMWG-modified frailty model to identify frail patients because of the importance of their identification for the treatment decision-making process.2 

Features associated with disease aggressiveness

The presence of extramedullary disease (EMD) or plasma cell leukemia (PCL), primary or secondary, is infrequent, but these are considered high-risk features not only because the plasma cells escape from the bone marrow environment but also because patients with EMD or PCL are difficult to treat, with poor outcomes with the current therapies (3-year survival rate is 35% for EMD3  and median OS is 12 months for PCL4 ).

Cytogenetic abnormalities

The IMWG currently recommends the detection of t(4;14), t(14;16) and del (17/17p) in selected plasma cells by interphase fluorescent in situ hybridization for the identification of high-risk patients.5  In newly diagnosed MM (NDMM), the presence of at least 1 high-risk cytogenetic abnormality (CA) is associated with a median OS of 24.5 months, significantly shorter than the 50.5 months observed when there are not any CAs (P  <  .001). This is far from complete and requires being updated.

The gain of the long arm of chromosome 1 (+1q) is a frequent CA observed in approximately 30% of NDMMs and associated with poor outcome. A retrospective study conducted in 201 patients with NDMM treated with RVd reported that patients harboring +1q had a shorter median progression-free survival (PFS; 41.9 months) and OS (not reached) compared with those without +1q (PFS of 65.1 months, P  =  .002 and OS not reached, P  =  .003). The negative impact on survival with +1q may be more profound if there is amplification of 1q, defined by the presence of 4 or more copies of chromosome 1q (median PFS of 25.1 months) or in association with other high-risk CAs (median PFS of 34.6 months). The poor prognosis of patients who have +1q with a coexisting deletion of chromosome 1p (del(1p)) has also been described.6 

Although it is well accepted that del(17p) is the CA with more prognostic impact in MM, some questions are under debate: (1) What is the optimal threshold to predict poor prognosis? (2) Is TP53 an optimal molecular target? (3) What about mono- or biallelic deletion and/or inactivation of TP53 through mutations? Patients with a “double-hit” biallelic inactivation of TP53 are at high risk, especially if this abnormality coexists with ISS 3 (1.5-year PFS of 33%) in ND patients.7  The Intergroupe Francophone du Myélome (IFM) has recently reported in a large series of patients with NDMM that the presence of isolated del(17p) was also associated with a poor outcome, although the poorest outcome was reported for the double-hit patients.8 

In summary, the optimal identification of high-risk MM based on CA is under construction, but in clinical practice, the CA recommended by the IMWG, together with abnormalities of chromosome 1 and mutational status of TP53, if possible, would be necessary to define the risk at baseline. At the moment of the relapse, it would be optimal to repeat these evaluations because of the clonal evolution and the potential acquisition of new CAs not detectable at baseline.

R-ISS

The ISS risk model, including albumin and β2-microglobulin levels, was improved with the incorporation of 2 well-known disease-related prognostic biomarkers, CA and serum lactate dehydrogenase (LDH) levels, which are associated with a higher proliferative activity, resulting in the R-ISS model.

The R-ISS emerged from a large series of patients with NDMM, and 3 subgroups were defined: R-ISS I (n  =  871), including ISS stage I, no high-risk CA, and normal LDH level with a 5-year OS rate of 82%; R-ISS III (n  =  295), including ISS stage III and high-risk CA or high LDH level with a 5-year OS of 40%; and R-ISS II (n  =  1894), including all the other possible combinations and with a 5-year OS of 62%.9  This staging system is still valid, although there are some limitations: (1) most patients were assigned to the R-ISS II, including those with high-risk CA but not ISS III, (2) some other high-risk CAs such as +1q were not included, and (3) other, more complex genomic abnormalities such as mutations or inactivation of TP53 were not considered.

Functional high-risk patients

In addition to the above high-risk features, how do we recognize those patients with no apparent high risk at diagnosis but who progress within the first 12 to 18 months after an optimal first line of therapy? These patients are functional high risk with poor prognosis, and further investigations are required to unravel if there is a clonal selection or just an inadequate evaluation at diagnosis.

If the identification of high-risk patients with MM is challenging, its management is not easy either. So far, only a few clinical trials have been specifically conducted in this population because the definition is heterogeneous. In addition, high-risk subgroups in clinical trials are quite small to generate solid recommendations.

In 2021, we know that poor prognosis with the presence of high-risk features can be at least improved or even abrogated by the achieving a deep and sustained response over time, which can most likely be obtained through the use of novel therapeutic options.10 

At least 3 large meta-analyses support the use of MRD for monitoring the response in MM because of its prognostic value. The most recent one included publications up to June 2019, showing that the achievement of undetectable MRD improved PFS (hazard ratio [HR], 0.33) and OS (HR, 0.45) in comparison with the presence of MRD. Moreover, its prognostic impact was sustained across the different subgroups, including those with some high-risk features such as elderly patients with NDMM, relapsed/refractory patients, or even the presence of high-risk CA.11 

Of note, the higher the sensitivity threshold for the MRD evaluation and the longer the sustained undetectable MRD over time, the higher the prognostic value.

In addition, the achievement of undetectable MRD can convert risk assessment in MM into something dynamic, and the high risk at baseline can be overcome when undetectable MRD is achieved. In the PETHEMA/GEM2012MENOS65 trial, 458 patients with NDMM had longitudinal assessment of MRD after 6 induction cycles with RVd, autologous transplantation, and 2 consolidation courses with RVd. The 3-year PFS rate for patients with R-ISS I, II, or III was comparable (95%, 94%, and 88%) if MRD was undetectable after treatment. By contrast, outcomes were progressively poor for patients with R-ISS I, II, and III when MRD was detectable, with a 3-year PFS of 62%, 53%, and 28%, respectively, and analogous results were observed when OS was considered. Similarly, outcome of patients with high-risk CA was abrogated when undetectable MRD was achieved.12 

One additional aspect needs to be incorporated in the MRD assessment: the MRD evaluation outside of the bone marrow through the use of functional imaging tools such as positron emission tomography/computerized tomography.13  Deauville scores to focal lesions less than 4 and bone marrow uptake showing the liver background (Deauville score <4) have been identified as the best cutoff to define positron emission tomography/computerized tomography negativity after therapy and complete metabolic response, as they have been described in at least 2 clinical trials, the FORTE and CASSIOPETT substudy of CASSIOPEIA trial.

Management of frail patients

The general approach described above is feasible for frail patients, but tolerability and quality of life are crucial to deliver treatments in order to reach depth responses. At least 1 clinical trial has been conducted in unfit and frail patients with NDMM according to the IMWG frailty index, using ixazomib and daratumumab plus very low dose of dexamethasone.14  Preliminary results are encouraging, with 1-year OS rates of 96% and 74% for unfit and frail patients, respectively. Subgroup analysis recently conducted in the phase 3 trials ALCYONE and MAIA have also shown how the addition of daratumumab to either bortezomib, melphalan, and prednisone (VMP) or Rd (lenalidomida and dexamethasone) has been able to significantly improve the outcome of frail patients compared with VMP or Rd alone, according to a modified IMWG frailty index (Table 2). In the relapsed-refractory setting, other subanalyses of phase 3 clinical trials have reported how carfilzomib at different doses and schedules or the combination of pomalidomide-dexamethasone plus isatuximab is feasible and able to improve the outcome of frail patients.15,16  Although this information is obtained from clinical trials, the good toxicity profile of all novel agents makes it possible to maintain therapy in the frail population.

Table 2.

Clinical studies ongoing in patients with high-risk features

Registration numberStudy designPopulation
ND high-risk MM 
NCT03104842 Isatuximab-KRd as induction, consolidation, and maintenance Transplant eligible or ineligible
del(17p) in ≥10% of purified cells and/or t(4;14) and/or >3 copies +1q21
ISS II or III 
NCT03756896 Carfilzomib, pomalidomide, and dexamethasone as maintenance after HDM-ASCT Transplant eligible achieving at least partial response
Presence of del(17p), t(4;14), t(14;16), t(14;20)
PCL at diagnosis 
NCT04025450 Chidamide (HDAC inhibitor)–lenalidomide, bortezomib, and dexamethasone as induction Transplant eligible and ineligible
Presence of del(17p), t(4;14), t(14;16), t(14;20)
R-ISS III
IgD/IgE
Extramedullary plasmacytomas
Peripheral plasma cells by flow cytometry ≥0.165% 
NCT02128230 Induction with melphalan 20–KTD-PACE followed by melphalan 80–KTd-PACE plus ASCT and KTD-PACE consolidation and KRd maintenance (1 year) and Kd (1 year) Transplant eligible

GEP70 risk score of ≥0.66 
NCT03549442 BCMA CAR-T + huCART19 in different schedules ISS III or R-ISS III or
Metaphase karyotype with >3 abnormalities except hyperdiploidy
Failure to achieve partial response or better to initial therapy based on PI and IMiD 
NCT04196491 BCMA CAR-T bb2121 (ide-cel) (150-800  ×  106) followed by lenalidomide as maintenance R-ISS III 
NCT04436029 Autologous CD8+ T cells expressing an anti-BCMA chimeric antigen receptor High-risk patients who completed pretransplant induction antimyeloma treatment 
NCT04133636 BCMA CAR-T JNJ-68284528 (cilta-cel) followed by lenalidomide maintenance Less than complete response after first-line treatment and transplant followed or not by consolidation 
NCT04133636 BCMA CAR-T JNJ-68284528 (cilta-cel) followed by lenalidomide and daratumumab maintenance Noneligible for transplant patients with ISS III 
Relapsed-refractory high-risk MM 
NCT03601078 BCMA CAR-T bb2121 (150-450  ×  106R-ISS III and
PD <18 months after the first-line treatment including induction, transplant, and lenalidomide maintenance
PD <18 months since date of start initial therapy, which must contain PI, IMiD, and dexamethasone
Less thanVGPR after induction, including PI, IMiD, and dexamethasone and transplant (between 70 and 110 days after transplant) 
NCT04133636 BCMA CAR-T JNJ-68284528 (cilta-cel) One prior line including PI, IMiD, and PD within the first 12 months after transplant or the first-line treatment for nontransplant eligible 
NCT03104270 Elotuzumab in combination with pomalidomide, carfilzomib, and dexamethasone More than 2 prior lines, including PI and IMiD and
del(17p), t(14;16), t(14:20)
PCL
Extramedullary disease
Doubling in levels of MM markers in the past 3 months
Refractoriness to their most recent lenalidomide-containing regimen and PI-based regimen
Renal failure with CrCl between 15 and 30  mL  ×  minute 
Registration numberStudy designPopulation
ND high-risk MM 
NCT03104842 Isatuximab-KRd as induction, consolidation, and maintenance Transplant eligible or ineligible
del(17p) in ≥10% of purified cells and/or t(4;14) and/or >3 copies +1q21
ISS II or III 
NCT03756896 Carfilzomib, pomalidomide, and dexamethasone as maintenance after HDM-ASCT Transplant eligible achieving at least partial response
Presence of del(17p), t(4;14), t(14;16), t(14;20)
PCL at diagnosis 
NCT04025450 Chidamide (HDAC inhibitor)–lenalidomide, bortezomib, and dexamethasone as induction Transplant eligible and ineligible
Presence of del(17p), t(4;14), t(14;16), t(14;20)
R-ISS III
IgD/IgE
Extramedullary plasmacytomas
Peripheral plasma cells by flow cytometry ≥0.165% 
NCT02128230 Induction with melphalan 20–KTD-PACE followed by melphalan 80–KTd-PACE plus ASCT and KTD-PACE consolidation and KRd maintenance (1 year) and Kd (1 year) Transplant eligible

GEP70 risk score of ≥0.66 
NCT03549442 BCMA CAR-T + huCART19 in different schedules ISS III or R-ISS III or
Metaphase karyotype with >3 abnormalities except hyperdiploidy
Failure to achieve partial response or better to initial therapy based on PI and IMiD 
NCT04196491 BCMA CAR-T bb2121 (ide-cel) (150-800  ×  106) followed by lenalidomide as maintenance R-ISS III 
NCT04436029 Autologous CD8+ T cells expressing an anti-BCMA chimeric antigen receptor High-risk patients who completed pretransplant induction antimyeloma treatment 
NCT04133636 BCMA CAR-T JNJ-68284528 (cilta-cel) followed by lenalidomide maintenance Less than complete response after first-line treatment and transplant followed or not by consolidation 
NCT04133636 BCMA CAR-T JNJ-68284528 (cilta-cel) followed by lenalidomide and daratumumab maintenance Noneligible for transplant patients with ISS III 
Relapsed-refractory high-risk MM 
NCT03601078 BCMA CAR-T bb2121 (150-450  ×  106R-ISS III and
PD <18 months after the first-line treatment including induction, transplant, and lenalidomide maintenance
PD <18 months since date of start initial therapy, which must contain PI, IMiD, and dexamethasone
Less thanVGPR after induction, including PI, IMiD, and dexamethasone and transplant (between 70 and 110 days after transplant) 
NCT04133636 BCMA CAR-T JNJ-68284528 (cilta-cel) One prior line including PI, IMiD, and PD within the first 12 months after transplant or the first-line treatment for nontransplant eligible 
NCT03104270 Elotuzumab in combination with pomalidomide, carfilzomib, and dexamethasone More than 2 prior lines, including PI and IMiD and
del(17p), t(14;16), t(14:20)
PCL
Extramedullary disease
Doubling in levels of MM markers in the past 3 months
Refractoriness to their most recent lenalidomide-containing regimen and PI-based regimen
Renal failure with CrCl between 15 and 30  mL  ×  minute 

cilta-cel, ciltacabtagene autoleucel; CrCL, creatinine clearance; GEP70, gene expression profiling-70; HDAC, histone deacetylase; ide-cel, idecabtagene vicleucel; IMiD, immunomodulatory drug; Kd, carfilzomib and dexamethasone; KRd, carfilzomib, lenalidomide and dexamethasone; KTD-PACE, carfilzomib, thalidomide, dexamethasone, cisplatin, doxorubicin, cyclophosphamide, and etoposide; PD, progression disease; VGPR, very good partial response.

Management of patients with high-risk CA or R-ISS III with approved drugs

Proteasome inhibitors (PIs), immunomodulatory drugs, and corticosteroids are the key treatment elements of patients with MM patients with high-risk CA. For transplant-eligible patients with NDMM, the question about bortezomib or carfilzomib as the optimal PI for high-risk patients remains under debate because the only phase 3 randomized trial comparing bortezomib with carfilzomib did not show any difference, but it did only include patients with t(4;14).17 

The use of moAbs (monoclonal antibodies) targeting SLAMF7 and CD38 also has been evaluated in this setting. Although the addition of elotuzumab showed no significant benefit when combined with RVd in the phase 2 SWOG-1211 study,18  the addition of daratumumab has been shown to improve the outcome of patients with NDMM and RRMM (relapsed refractory multiple myeloma) with high-risk CA in a recent meta-analysis.19  HDM-ASCT continues to be the standard of care in high-risk NDMM because of its capacity to achieve a higher undetectable MRD rate, and tandem transplant is even considered for this population based on the positive data from the EMN02 trial, confirmed in the STAMINA trial at least in terms of PFS.20,21  However, tandem transplant may not be necessary with the introduction of moABs, especially with the introduction of cell therapy. Maintenance with lenalidomide is the standard of care to improve the outcome of high-risk patients compared with observation, but it should be improved through the addition of PIs or moABs, with preliminary positive data.22,23 

In the nontransplant-eligible, high-risk patients with NDMM, daratumumab, lenalidomide, and dexamethasone would be the first choice based on the results reported in the MAIA trial, with a median PFS of 45.3 months compared with 29.6 months in the Rd arm (HR, 0.57).24 

In the RR (relapse or refractory) setting, the same approach is valid, and the combination of choice for patients with high-risk CA would be those with the higher likely probability of achieving undetectable MRD (Table 2).

Of note, the novel drug melflufen flufenamide has shown promising efficacy in 45 patients with EMD (extramedullary disease) included in the HORIZON trial, with an overall response of 24% vs 30% in patients without EMD.25  Selinexor also may have a role in treating patients with del(17p) based on its mechanism of action and available evidence in some clinical trials.26  Belantamb mafodotin, a BCMA-conjugated moAb, produced a response rate of 33%, which is similar to that reported in patients with high-risk cytogenetics.27  Further studies are required to confirm this efficacy. Table 2 shows the efficacy reported in patients with high-risk CA in the most relevant clinical trials in patients with NDMM and RRMM.

Management of functional high-risk patients

Although no specific trials were performed until very recently, new trials with BCMA-targeted CAR-T cells have focused on this subgroup of patients (Table 3). Some subgroup analysis in phase 3 trials focused on early relapses, defined as those occurring within the first 12 to 18 months after the previous therapy, showing that the addition of carfilzomib to Rd or daratumumab to Rd vs Rd in the ASPIRE and POLLUX trials resulted in a significant benefit for the triple combination compared with Rd. A similar effect has been recently reported with the addition of daratumumab to bortezomib or carfilzomib28-31  (Table 2).

Table 3.

Efficacy of current treatment approaches for patients with NDMM with high-risk features (defined as del(17p), t(4;14), or t(14;16))

Clinical trialTransplant-eligible NDMM
SWOG-1211CassiopeiaForteGriffin
Population High risk* ITT High risk ITT High risk ITT High risk 
Treatment EloVRd vs VRd DVTd vs VTD DVTd vs VTD KRd-T/KRd12 KRd-T/KRd12 DRVd vs RVd DRVd vs RVd 
PFS (m)/HR 31 vs 34/0.96 NR/0.47 NR/0.67 NR/0.64 NR/0.51 NR/NA NR/NA 
Clinical trialTransplant-ineligible NDMM
SWOGALCYONEMAIA
Population ITT High risk ITT High risk Frail patients ITT High risk Frail patients 
Treatment VRd- > Rd vs Rd VRd- > Rd vs Rd DVMP vs VMP DVMP vs VMP DVMP vs VMP DRd vs Rd DRd vs Rd DRd vs Rd 
PFS (m)/HR 43 vs 30/0.74 38 vs 16/NA 36 vs 18/0.50 18 vs 18/0.78 33 vs 19/0.51 NR vs 34/0.54 45 vs 29/0.57 NR vs 30/0.62 
Clinical trialRelapsed-refractory MM
POLLUXASPIREELOQUENT-2TOURMALINE-MM1
Population ITT High risk Early relapse ITT High risk Early relapse ITT High risk ITT High risk 
Treatment DRd vs Rd DRd vs Rd KRd vs Rd KRd vs Rd EloRd vs Rd EloRd vs Rd IRd vs Rd IRd vs Rd 
PFS (m)/HR 44.5 vs 17.5/0.44 26.8 vs 8.3/0.37 0.38 26.3 vs 17.3/0.69 23 vs 13.9/0.7 21.4 vs 10.7/0.7 19.4 vs 14.9/0.70 NA/0.72(del17p)
NA/0.56 (t(4;14) 
20.6 vs 14.7/0.74 21.4 vs 9.7/0.54 
Clinical trialCASTORENDEAVORCANDORIKEMA
Population ITT High risk ITT High risk ITT High risk Early relapse ITT High risk 
Treatment DVd vs Vd DVd vs Vd Kd vs Vd Kd vs Vd DKd vs Kd DKd vs Kd IsaKd vs Kd IsaKd vs Kd 
PFS (m)/HR 16.7 vs 7.1/0.31 12.6 vs 6.2/0.41 18.7 vs 9.4/0.53 8.8 vs 6.0/0.7 28.6 vs 15.9/0.59 15.6 vs 5.6/0.49 CRrate 28 vs 3% NR vs 19.1/0.53 NA/0.72 
Clinical trialOPTIMISMMBOSTONICARIAELOQUENT-3
Population ITT High risk ITT High risk ITT High risk ITT High risk 
Treatment PVd vs Vd PVd vs Vd SVd vs Vd SVd vs Vd IsaPd vs Pd IsaPd vs Pd EloPd vs Pd EloPd vs Pd 
PFS (m)/HR 11.2 vs 7.1/0.61 NA/0.56 11.2 vs 5.8/0.61 NA/0.67 11.5 vs 6.4/0.59 NA/0.66 10.3 vs 4.7/0.54 0.52 
Clinical trialSTORMHORIZONDREAMM-2KARMMA-1
Population ITT High risk ITT High risk ITT High risk ITT High risk 
Treatment Sd Sd Melflufen-dex Melflufen-dex Belamaf Belamaf Ide-cel Ide-cel 
PFS (m)/HR 3.7 3.3* and 4.6* 4.2 3.0 3.9 2.1 8.8 10.4 
Clinical trialTransplant-eligible NDMM
SWOG-1211CassiopeiaForteGriffin
Population High risk* ITT High risk ITT High risk ITT High risk 
Treatment EloVRd vs VRd DVTd vs VTD DVTd vs VTD KRd-T/KRd12 KRd-T/KRd12 DRVd vs RVd DRVd vs RVd 
PFS (m)/HR 31 vs 34/0.96 NR/0.47 NR/0.67 NR/0.64 NR/0.51 NR/NA NR/NA 
Clinical trialTransplant-ineligible NDMM
SWOGALCYONEMAIA
Population ITT High risk ITT High risk Frail patients ITT High risk Frail patients 
Treatment VRd- > Rd vs Rd VRd- > Rd vs Rd DVMP vs VMP DVMP vs VMP DVMP vs VMP DRd vs Rd DRd vs Rd DRd vs Rd 
PFS (m)/HR 43 vs 30/0.74 38 vs 16/NA 36 vs 18/0.50 18 vs 18/0.78 33 vs 19/0.51 NR vs 34/0.54 45 vs 29/0.57 NR vs 30/0.62 
Clinical trialRelapsed-refractory MM
POLLUXASPIREELOQUENT-2TOURMALINE-MM1
Population ITT High risk Early relapse ITT High risk Early relapse ITT High risk ITT High risk 
Treatment DRd vs Rd DRd vs Rd KRd vs Rd KRd vs Rd EloRd vs Rd EloRd vs Rd IRd vs Rd IRd vs Rd 
PFS (m)/HR 44.5 vs 17.5/0.44 26.8 vs 8.3/0.37 0.38 26.3 vs 17.3/0.69 23 vs 13.9/0.7 21.4 vs 10.7/0.7 19.4 vs 14.9/0.70 NA/0.72(del17p)
NA/0.56 (t(4;14) 
20.6 vs 14.7/0.74 21.4 vs 9.7/0.54 
Clinical trialCASTORENDEAVORCANDORIKEMA
Population ITT High risk ITT High risk ITT High risk Early relapse ITT High risk 
Treatment DVd vs Vd DVd vs Vd Kd vs Vd Kd vs Vd DKd vs Kd DKd vs Kd IsaKd vs Kd IsaKd vs Kd 
PFS (m)/HR 16.7 vs 7.1/0.31 12.6 vs 6.2/0.41 18.7 vs 9.4/0.53 8.8 vs 6.0/0.7 28.6 vs 15.9/0.59 15.6 vs 5.6/0.49 CRrate 28 vs 3% NR vs 19.1/0.53 NA/0.72 
Clinical trialOPTIMISMMBOSTONICARIAELOQUENT-3
Population ITT High risk ITT High risk ITT High risk ITT High risk 
Treatment PVd vs Vd PVd vs Vd SVd vs Vd SVd vs Vd IsaPd vs Pd IsaPd vs Pd EloPd vs Pd EloPd vs Pd 
PFS (m)/HR 11.2 vs 7.1/0.61 NA/0.56 11.2 vs 5.8/0.61 NA/0.67 11.5 vs 6.4/0.59 NA/0.66 10.3 vs 4.7/0.54 0.52 
Clinical trialSTORMHORIZONDREAMM-2KARMMA-1
Population ITT High risk ITT High risk ITT High risk ITT High risk 
Treatment Sd Sd Melflufen-dex Melflufen-dex Belamaf Belamaf Ide-cel Ide-cel 
PFS (m)/HR 3.7 3.3* and 4.6* 4.2 3.0 3.9 2.1 8.8 10.4 
*

High-risk definition: gene expression profiling high risk, t(14;16), t(14;20), del(17p), amp1q21, plasma cell leukemia, elevated serum LDH (2  ×  upper limit of normal).

Belamaf, belantamaf mafodotin; dex, dexamethasone; DKd, daratumumab, carfilzomib, and dexamethasone; DRd, daratumumab, lenalidomide, and dexamethasone; DRVd, daratumumab, lenalidomide, bortezomib, and dexamethasone; DVd, daratumumab, bortezomib, and dexamethasone; DVMP, daratumumab, bortezomib, melphalan, and prednisone; DVTd, daratumumab, bortezomib, thalidomide, and dexamethasone; Elo, elotuzumab; EloPd, elotuzumab, pomalidomide, and dexamethasone; EloRd, elotuzumab, lenalidomide, and dexamethasone; Ide-cel, idecabtagene vicleucel; IRd, ixazomib, lenalidomide, and dexamethasone; IsaKd, isatuximab, carfilzomib, and dexamethasone; IsaPd, isatuximab, pomalidomide, and dexamethasone; ITT, intention-to-treat population; Kd, carfilzomib and dexamethasone; KRd, carfilzomib, lenalidomide, and dexamethasone; KRd-T, carfilzomib, lenalidomide, and dexamethasone followed by transplant; KRd 12, KRd for 12 cycles; m, months; NA, not available; NR, not reached; PVd, pomalidomide, bortezomib, and dexamethasone; Rd, lenalidomide and dexamethasone; RVd, lenalidomide, bortezomib, and dexamethasone; Sd, selinexor and dexamethasone; SVd, selinexor, bortezomib, and dexamethasone; VTd, bortezomib, thalidomide, and dexamethasone.

Management of high-risk patients with CAR-T cell therapy

BCMA is an attractive and extensively studied target for immunotherapy in MM. BCMA-targeting CAR-T cells have demonstrated fast, high, and deep responses in patients with RRMM. The sample sizes across the different trials are so far rather small but have included a great proportion of patients with high-risk features, such as high-risk CA, R-ISS III, EMD, or high tumor burden.32,33  Ide-cel (idecabtagene vicleucel), indeed, already has been approved by US Food and Drug Administration for RRMM after at least 4 rounds of therapy, including PIs, immunomodulatory drugs, and anti-CD38, and has been evaluated in 128 patients with RRMM after a median of 6 prior lines (84% triple refractory). The ORR (overall response rate) was 73%, including a complete remission rate of 33% and a median PFS of 8.8 months. These efficacy data were sustained in patients with high-risk features, such as EMD (n  =  50), high-risk CA (n  =  45), or high tumor burden (n  =  65)34  (Table 2). Many other BCMA-targeted CAR-T cells are under investigation, and some clinical trials are focused in patients with high-risk features (Table 3). If results are positive, CAR-T cell therapy will rapidly move as the first choice in patients with high-risk features.

Beyond BCMA-targeted CAR-T cells, there are other therapeutic options, such as bispecific moABs targeting not only BCMA but also GPRC5D, FcRH5, and others, under evaluation in patients with RRMM, and their efficacy will be also evaluated in patients with high-risk features.

In summary, in 2021, the identification of high-risk patients with MM continues being an unmet medical need, and the definition should be revisited. Genomics will help us to improve the identification of these patients, as well as the therapeutic advances, to find the best option for them. Considering the achievement of undetectable and sustained MRD can abrogate the poor prognosis of high-risk features, the management of these patients should be response adapted and determined from exposition to sequential treatments based on drugs with a new and different mechanism of action. International effort and research in this regard are required.

María-Victoria Mateos: has received honoraria derived from lectures and advisory boards from Janssen, BMS-Celgene, Amgen, Takeda, Abbvie, Sanofi, Oncopeptides, Adaptive, Roche, Pfizer, Regeneron, GSK, Bluebird Bio, and Sea-Gen.

Borja Puertas Martínez: no competing financial interests to declare.

Verónica González-Calle: has received honoraria from Janssen and Celgene, received research funding from Janssen, and provided consulting or an advisory role for Prothena and Janssen.

María-Victoria Mateos: nothing to disclose.

Borja Puertas Martínez: nothing to disclose.

Verónica González-Calle: nothing to disclose.

1.
Palumbo
A
,
Bringhen
S
,
Mateos
M-V
, et al.
Geriatric assessment predicts survival and toxicities in elderly myeloma patients: an International Myeloma Working Group report
.
Blood
.
2015
;
125
(
13
):
2068
-
2074
.
doi:10.1182/blood-2014-12-615187
.
2.
Facon
T
,
Dimopoulos
MA
,
Meuleman
N
, et al.
A simplified frailty scale predicts outcomes in transplant-ineligible patients with newly diagnosed multiple myeloma treated in the FIRST (MM-020) trial
.
Leukemia
.
2020
;
34
(
1
):
224
-
233
.
doi:10.1038/s41375-019-0539-0
.
3.
Usmani
SZ
,
Heuck
C
,
Mitchell
A
, et al.
Extramedullary disease portends poor prognosis in multiple myeloma and is over-represented in high-risk disease even in the era of novel agents
.
Haematologica
.
2012
;
97
(
11
):
1761
1767
.
doi:10.3324/haematol.2012.065698
.
4.
Gonsalves
WI
,
Rajkumar
SV
,
Go
RS
, et al.
Trends in survival of patients with primary plasma cell leukemia: a population-based analysis
.
Blood
.
2014
;
124
(
6
):
907
-
912
.
doi:10.1182/blood-2014-03-565051
.
5.
Sonneveld
P
,
Avet-Loiseau
H
,
Lonial
S
, et al.
Treatment of multiple myeloma with high-risk cytogenetics: a consensus of the International Myeloma Working Group
.
Blood
.
2016
;
127
(
24
):
2955
-
2962
.
doi:10.1182/blood-2016-01-631200
.
6.
Schmidt
TM
,
Barwick
BG
,
Joseph
N
, et al.
Gain of chromosome 1q is associated with early progression in multiple myeloma patients treated with lenalidomide, bortezomib, and dexamethasone
.
Blood Cancer J
.
2019
;
9
(
12
):
94
.
doi:10.1038/s41408-019-0254-0
.
7.
Corre
J
,
Munshi
NC
,
Avet-Loiseau
H
.
Risk factors in multiple myeloma: is it time for a revision?
Blood
.
2021
;
137
(
1
):
16
-
19
.
doi:10.1182/blood.2019004309
.
8.
Corre
J
,
Perrot
A
,
Caillot
D
, et al.
del(17p) without TP53 mutation confers a poor prognosis in intensively treated newly diagnosed patients with multiple myeloma
.
Blood
.
2021
;
137
(
9
):
1192
-
1195
.
doi:10.1182/blood.2020008346
.
9.
Palumbo
A
,
Avet-Loiseau
H
,
Oliva
S
, et al.
Revised international staging system for multiple myeloma: a report from International Myeloma Working Group
.
J Clin Oncol
.
2015
;
33
(
26
):
2863
-
2869
.
doi:10.1200/JCO.2015.61.2267
.
10.
Burgos
L
,
Puig
N
,
Cedena
M-T
, et al.
Measurable residual disease in multiple myeloma: ready for clinical practice?
J Hematol Oncol
.
2020
;
13
(
1
):
82
.
doi:10.1186/s13045-020-00911-4
.
11.
Munshi
NC
,
Avet-Loiseau
H
,
Anderson
KC
, et al.
A large meta-analysis establishes the role of MRD negativity in long-term survival outcomes in patients with multiple myeloma
.
Blood Adv
.
2020
;
4
(
23
):
5988
-
5999
.
doi:10.1182/bloodadvances.2020002827
.
12.
Paiva
B
,
Puig
N
,
Cedena
M-T
, et al
;
GEM (Grupo Español de Mieloma)/PETHEMA (Programa Para el Estudio de la Terapéutica en Hemopatías Malignas) Cooperative Study Group
.
Measurable residual disease by next-generation flow cytometry in multiple myeloma
.
J Clin Oncol
.
2020
;
38
(
8
):
784
-
792
.
doi:10.1200/JCO.19.01231
.
13.
Zamagni
E
,
Nanni
C
,
Dozza
L
, et al.
Standardization of 18F-FDG-PET/CT according to Deauville criteria for metabolic complete response definition in newly diagnosed multiple myeloma
.
J Clin Oncol
.
2021
;
39
(
2
):
116
-
125
.
doi:10.1200/JCO.20.00386
.
14.
Stege
CAM
,
Nasserinejad
K
,
van der Spek
E
, et al.
Efficacy and tolerability of ixazomib, daratumumab and low dose dexamethasone (Ixa Dara dex) in unfit and frail newly diagnosed multiple myeloma (NDMM) patients; results of the interim efficacy analysis of the phase II HOVON 143 study
.
Blood
.
2019
;
134
(
suppl 1
):
695
-
695
.
doi:10.1182/blood-2019-121694
.
15.
Facon
T
,
Niesvizky
R
,
Mateos
M-V
, et al.
Efficacy and safety of carfilzomib-based regimens in frail patients with relapsed and/or refractory multiple myeloma
.
Blood Adv
.
2020
;
4
(
21
):
5449
-
5459
.
doi:10.1182/bloodadvances.2020001965
.
16.
Schjesvold
FH
,
Richardson
PG
,
Attal
M
, et al.
Efficacy of isatuximab with pomalidomide and dexamethasone in elderly patients with relapsed/refractory multiple myeloma: ICARIA-MM subgroup analysis
.
Blood
.
2019
;
134
(
suppl 1
):
1893
-
1893
.
doi:10.1182/blood-2019-128010
.
17.
Kumar
SK
,
Jacobus
SJ
,
Cohen
AD
, et al.
Carfilzomib or bortezomib in combination with lenalidomide and dexamethasone for patients with newly diagnosed multiple myeloma without intention for immediate autologous stem-cell transplantation (ENDURANCE): a multicentre, open-label, phase 3, randomised, controlled trial
.
Lancet Oncol
.
2020
;
21
(
10
):
1317
-
1330
.
doi:10.1016/S1470-2045(20)30452-6
.
18.
Usmani
SZ
,
Hoering
A
,
Ailawadhi
S
, et al
;
SWOG1211 Trial Investigators
.
Bortezomib, lenalidomide, and dexamethasone with or without elotuzumab in patients with untreated, high-risk multiple myeloma (SWOG-1211): primary analysis of a randomised, phase 2 trial
.
Lancet Haematol
.
2021
;
8
(
1
):
e45
e54
.
doi:10.1016/S2352-3026(20)30354-9
.
19.
Giri
S
,
Grimshaw
A
,
Bal
S
, et al.
Evaluation of daratumumab for the treatment of multiple myeloma in patients with high-risk cytogenetic factors: a systematic review and meta-analysis
.
JAMA Oncol
.
2020
;
6
(
11
):
1759
-
1765
.
doi:10.1001/jamaoncol.2020.4338
.
20.
Cavo
M
,
Gay
F
,
Beksac
M
, et al.
Autologous haematopoietic stem-cell transplantation versus bortezomib-melphalan-prednisone, with or without bortezomib-lenalidomide-dexamethasone consolidation therapy, and lenalidomide maintenance for newly diagnosed multiple myeloma (EMN02/HO95): a multicentre, randomised, open-label, phase 3 study
.
Lancet Haematol
.
2020
;
7
(
6
):
e456
-
e468
.
doi:10.1016/S2352-3026(20)30099-5
.
21.
Hari
P
,
Pasquini
MC
,
Stadtmauer
EA
, et al.
Long-term follow-up of BMT CTN 0702 (STaMINA) of postautologous hematopoietic cell transplantation (autoHCT) strategies in the upfront treatment of multiple myeloma (MM)
.
J Clin Oncol
.
2020
;
38
(
15, suppl
):
8506
.
doi:10.1200/JCO.2020.38.15_suppl.8506
22.
Voorhees
PM
,
Kaufman
JL
,
Laubach
J
, et al.
Daratumumab, lenalidomide, bortezomib, and dexamethasone for transplant-eligible newly diagnosed multiple myeloma: the GRIFFIN trial
.
Blood
.
2020
;
136
(
8
):
936
-
945
.
doi:10.1182/blood.2020005288
.
23.
Gay
F
,
Musto
P
,
Rota Scalabrini
D
, et al.
Survival analysis of newly diagnosed transplant-eligible multiple myeloma patients in the randomized Forte trial
.
Blood
.
2020
;
136
(
suppl 1
):
35
-
37
.
doi:10.1182/blood-2020-136907
.
24.
Facon
T
,
Kumar
S
,
Plesner
T
, et al.
MAIA Trial Investigators
.
Daratumumab plus lenalidomide and dexamethasone for untreated myeloma
.
N Engl J Med
.
2019
;
380
(
22
):
2104
-
2115
.
doi:10.1056/NEJMoa1817249
.
25.
Richardson
PG
,
Oriol
A
,
Larocca
A
, et al.
HORIZON (OP-106) Investigators. Melflufen and dexamethasone in heavily pretreated relapsed and refractory multiple myeloma
.
J Clin Oncol
.
2021
;
39
(
7
):
757
-
767
.
doi:10.1200/JCO.20.02259
.
26.
Chari
A
,
Vogl
DT
,
Gavriatopoulou
M
, et al.
Oral selinexor-dexamethasone for triple-class refractory multiple myeloma
.
N Engl J Med
.
2019
;
381
(
8
):
727
738
.
doi:10.1056/NEJMoa1903455
.
27.
Cohen
AD
,
Trudel
S
,
Lonial
S
, et al.
DREAMM-2: single-agent belantamab mafodotin (GSK2857916) in patients with relapsed/refractory multiple myeloma (RRMM) and high-risk (HR) cytogenetics
.
J Clin Oncol
.
2020
;
38
(
15, suppl
):
8541
.
doi:10.13039/100004330
.
28.
Mateos
M-V
,
Goldschmidt
H
,
San-Miguel
J
, et al.
Carfilzomib in relapsed or refractory multiple myeloma patients with early or late relapse following prior therapy: a subgroup analysis of the randomized phase 3 ASPIRE and ENDEAVOR trials
.
Hematol Oncol
.
2018
;
36
(
2
):
463
-
470
.
doi:10.1002/hon.2499
.
29.
Dimopoulos
MA
,
San-Miguel
J
,
Belch
A
, et al.
Daratumumab plus lenalidomide and dexamethasone versus lenalidomide and dexamethasone in relapsed or refractory multiple myeloma: updated analysis of POLLUX
.
Haematologica
.
2018
;
103
(
12
):
2088
-
2096
.
doi:10.3324/haematol.2018.194282
.
30.
Spencer
A
,
Lentzsch
S
,
Weisel
K
, et al.
Daratumumab plus bortezomib and dexamethasone versus bortezomib and dexamethasone in relapsed or refractory multiple myeloma: updated analysis of CASTOR
.
Haematologica
.
2018
;
103
(
12
):
2079
-
2087
.
doi:10.3324/haematol.2018.194118
.
31.
Weisel
K
,
Geils
GF
,
Karlin
L
, et al.
Carfilzomib, dexamethasone, and daratumumab versus carfilzomib and dexamethasone in relapsed or refractory multiple myeloma: subgroup analysis of the phase 3 candor study in patients with early or late relapse
.
Blood
.
2020
;
136
(
suppl 1
):
37
-
38
.
doi:10.1182/blood-2020-133908
32.
Munshi
NC
,
Anderson
LD
Jr
,
Shah
N
, et al.
Idecabtagene vicleucel in relapsed and refractory multiple myeloma
.
N Engl J Med
.
2021
;
384
(
8
):
705
716
.
doi:10.1056/NEJMoa2024850
.
33.
Madduri
D
,
Berdeja
JG
,
Usmani
SZ
, et al.
CARTITUDE-1: phase 1b/2 study of ciltacabtagene autoleucel, a B-cell maturation antigen-directed chimeric antigen receptor T cell therapy, in relapsed/refractory multiple myeloma
.
Blood
.
2020
;
136
(
suppl 1
):
22
-
25
.
doi:10.1182/blood-2020-136307
.
34.
Raje
NS
,
Siegel
DS
,
Jagannath
S
, et al.
Idecabtagene vicleucel (ide-cel, bb2121) in relapsed and refractory multiple myeloma: analyses of high-risk subgroups in the KarMMa study
.
Blood
.
2020
;
136
(
suppl 1
):
37
-
38
.
doi:10.1182/blood-2020-134319
.