Review Article

Split Viewer

Blood Res 2020; 55(S1):

Published online July 31, 2020

https://doi.org/10.5045/br.2020.S007

© The Korean Society of Hematology

Frontline therapy for newly diagnosed patients with multiple myeloma

Sung-Hoon Jung1, Jae-Cheol Jo2, Ga-Young Song1, Seo-Yeon Ahn1, Deok-Hwan Yang1, Jae-Sook Ahn1, Hyeoung-Joon Kim1, Je-Jung Lee1

1Department of Hematology-Oncology, Chonnam National University Hwasun Hospital, Hwasun, 2Department of Hematology and Oncology, Ulsan University Hospital, University of Ulsan College of Medicine, Ulsan, Korea

Correspondence to : Je-Jung Lee, M.D., Ph.D.
Department of Hematology-Oncology, Chonnam National University Hwasun Hospital, 322 Seoyangro, Hwasun 58128, Korea
E-mail: drjejung@chonnam.ac.kr

Received: November 13, 2019; Revised: January 22, 2020; Accepted: January 30, 2020

This is an Open Access article distributed unAcute myeloid leukemia, New FDA approvalsder the terms of the Creative Commons Attribution Non-Commercial License (http://creativecommons.org/licenses/by-nc/4.0) which permits unrestricted non-commercial use, distribution, and reproduction in any medium, provided the original work is properly cited.

Since the introduction of an alkylator to the treatment of multiple myeloma (MM), new effective agents have been developed, such as immunomodulatory drugs including thalidomide, lenalidomide, and pomalidomide; proteasome inhibitors including bortezomib, carfilzomib, and ixazomib; monoclonal antibodies including daratumumab and elotuzumab; and deacetylase inhibitors including panobinostat. Numerous regimens with these new agents have been developed and they have contributed in improving survival outcomes in MM patients. In addition, the recommended therapies for newly diagnosed MM change every year based on the results of clinical trials. This review will discusses the appropriate induction therapies based on recent clinical trials for patients with newly diagnosed MM.

Keywords Multiple myeloma, Induction therapy, Prognosis

Multiple myeloma (MM) is a clonal B-cell malignancy characterized by anemia, hypercalcemia, renal insufficiency, osteolytic bone lesions, and decreased immune function due to aberrant proliferation of malignant plasma cells in the bone marrow [1]. MM is the second-most common hematologic malignancy and it accounts for 10% of all hematologic malignancies in the United States [2]. In Korea, the incidence of MM is lower than that in Western countries, but is increasing rapidly over the last three decades. According to the 2012 Korean data on national cancer statistics, the age-standardized incidence rate of MM in 2012 was 10 times higher than that 20 years ago [3, 4]. This increase in MM incidence in Korea is presumed to be due to aging, exposure to environmental risk and improved early detection by the expansion of health insurance, relevant education of health providers, and public awareness of this disease.

Treatment options for MM have rapidly expanded in the recent decades. In the 1980s, induction therapy with alkylating agents, such as anthracyclines and corticosteroids, as well as high-dose chemotherapy followed by autologous stem cell transplantation (HDT/ASCT) were the main treatment approaches for MM [5, 6]. However, these treatment approaches have markedly changed since the introduction of several more effective novel agents, such as proteasome inhibitors, including bortezomib, carfilzomib, and ixazomib, and immunomodulatory drugs, including thalidomide, lenalidomide, and pomalidomide. Treatment with these agents improves response rates in relapsed or refractory MM. Furthermore, their use in induction therapy resulted in considerable improvement of outcomes and extended overall survival (OS) times [7, 8]. Numerous regimens with these agents have been developed and the result of clinical trials with new regimens are reported each year. Furthermore, the recent development of monoclonal antibodies and histone deacetylase inhibitor for MM have further expanded the treatment landscape by improving deep response rate and response duration.

The purpose of this review was to suggest the optimal combination for induction therapy based on recent clinical data for newly diagnosed patients with MM.

Risk stratification

After being diagnosed with MM, patients must be evaluated for risk classification. Although the introduction of effective new agents for MM has improved survival outcomes, response to treatment is highly variable. Some patients may survive for more than 10 years, whereas others with an aggressive course may survive for less than 2 years. In general, high-risk MM is defined as the disease subgroup with a median OS of less than 3 years in transplant-eligible patients and less than 2 years in transplant-ineligible patients [9]. Identification of this subgroup at diagnosis would be beneficial to the personalized and intensive treatment approach from the time of induction therapy.

There have been numerous studies of accurate prognostic classification, and various prognostic staging systems have been proposed. Among them, the International Staging System (ISS) proposed by Greipp et al. [10] in 2005 was a preferred staging system. The ISS was developed using survival data from 10,750 patients from 1981 to 2002. However, several data of patients treated with novel agents were included in the analysis for ISS, and there was a debate for the prognosis value of ISS in the era of novel agents. In 2015, the Revised ISS (R-ISS) was suggested by the International Myeloma Working Group to improve risk stratification by adding cytogenetic abnormalities and serum lactate dehydrogenase (LDH) to the original ISS [11]. Cytogenetic abnormalities in R-ISS included del(17p), t(4;14), and/or t(14;16). Using these parameters, the prognostic power of the R-ISS was improved compared to that of the ISS, and showed predictive value independent of patients’ age, performance of ASCT, and treatment received. In addition to the cytogenetic abnormalities included in the R-ISS, t(14;20), gains/amplification of 1q21, and del(1p) are also considered high-risk features [9, 12]. Accumulation of these cytogenetic abnormalities is associated with poor prognosis. The study of 1,069 MM patients treated in the Medical Research Council (MRC) Myeloma IX trial firstly showed the poor prognosis of patients who had had any two of the following abnormalities: 1) adverse translocations t(4;14), t(14;16), and t(14;20); 2) gain(1q); 3) del(17p) [13]. In addition, recent genome-wide analysis of the large data set of molecular and clinical data from newly diagnosed MM, ‘double hit’ myeloma was defined as the presence of ≥1 of the following two adverse genetic factors: a) bi-allelic TP53 inactivation and b) amplification (≥4 copies) of CKS1B (1Q21). It constituted 6.1% of the patients and was associated with median progression-free survival (PFS) of 15.4 months and median OS of 20.7 months despite modern therapies [14]. Analysis of molecular subtype by gene expression profiling (GEP) for MM is expected to provide additional prognostic information and to help making therapeutic decisions. Several studies have reported that 15-gene, 70-gene, or 92-gene models based on GEP had prognostic significance in MM [15-17].

Imaging at diagnosis, such as MRI and 18F-fluorodeoxyglucose (18F-FDG) positron emission tomography/computed tomography (PET/CT), has been shown to be associated with prognosis. The diffuse pattern of marrow infiltration in MRI was associated with inferior survival than that with other MRI patterns, such as focal, variegated, and normal MRI patterns [18]. In addition, the number of FDG-avid focal lesions, presence of extramedullary disease, and maximum standardized uptake value have been reported as reliable prognostic 18F-FDG PET/CT parameters in MM [19-21].

Assessment of eligibility for ASCT

The Intergroupe Francophone du Myelome (IFM) study was the first randomized trial showing the superiority of high-dose melphalan and total body irradiation followed by ASCT over the conventional chemotherapy in terms of response rate, event-free survival, and OS [22]. Subsequent randomized trials have also shown that HDT/ASCT, compare to conventional chemotherapy, improved response rates and survival [6, 23]. Although there are some debates regarding the role of HDT/ASCT in MM with the emergence of new and more effective agents, upfront ASCT did lead to better response and longer PFS in a recent randomized phase 3 EMN02/HO95 trial [HR, 0.76; 95% confidence interval (CI), 0.64–0.91; P=0.002] [24]. In addition, a meta-analysis of three large phase 3 randomized controlled trials showed the superiority of upfront ASCT over novel agents [25]. Therefore, assessment of eligibility for ASCT at diagnosis is important to decide treatment strategy for MM. Transplant-eligible patients typically receive four to six cycles of induction therapy, followed by HDT/ASCT and then maintenance therapy until progression.

Age, generally age of 65 years, was considered a main factor in determining ASCT. However, recently, HDT/ASCT has been extended to older patients and has shown comparable outcome to that in younger patients [26, 27]. It is necessary to determine whether ASCT will proceed by evaluating frailty parameters, including performance status, organ function, and comorbidities, rather than age alone.

Transplant-eligible patients

The main result of recent clinical trials in transplant-eligible patients with newly diagnosed MM are summarized in Table 1. A combination regimen of three drugs including a proteasome inhibitor is preferred for induction therapy in MM. Bortezomib, thalidomide, and dexamethasone (VTD) regimen is a preferred regimen and has been evaluated in three randomized phase III trials [28-30]. Although the dose intensity and treatment cycle of VTD were different in three randomized trials, six cycles of VTD prior to ASCT showed superior CR rate than that shown by three cycles of VTD (35% vs. 19%). In the IFM trial, patients received four cycles of VTD with lower bortezomib dose of 1.0 mg/m2 and CR rates prior to ASCT of only 13%. However, the incidence of grade 3 or 4 peripheral neuropathy was lower with reduced VTD than with four cycles of VTD or six cycles of VTD (3% vs. 10% vs. 14%). Therefore, the dose intensity and treatment duration of VTD regimen were important to achieve deep response, but increased dosing and treatment cycle were also associated with the occurrence of peripheral neuropathy.

Table 1 Summary of major recent clinical trials in transplant-eligible patients with newly diagnosed multiple myeloma.

Ref.RegimenCycle of inductionNCR rate prior to transplantMedian PFSMedian OS
Cavo et al. [28]VTD vs.323619%3-year PFS; 68%3-year OS; 86%
TD2385%vs. 56%vs. 84%
Rosiñol et al. [29]VTD vs.613035%56.2 mo4-year OS; 74%
TD vs.12714%28.2 movs. 70%
VBMCP/VBAD/B1290.2135.3 movs. 65%
Moreau et al. [31]VTD vs.416913%NANA
VCD1698.9%
Rosiñol et al. [33]VRD645833.4%NANA
Moreau et al. [37]D-VTD vs.454339%18 mo PFS; 93%NA
VTD54226%vs. 85%

Abbreviations: CR, complete response; D-VTD; daratumumab, bortezomib, thalidomide, and dexamethasone; NA, not evaluable; OS, overall survival; PFS, progression-free survival; Ref, reference; TD, thalidomide and dexamethasone; VBMCP/VBAD, vincristine, VCD, bortezomib, cyclophosphamide, and dexamethasone; VRD, bortezomib, lenalidomide, and dexamethasone; VTD, bortezomib, thalidomide, and dexamethasone.



Another effective regimen was bortezomib, cyclophosphamide, and dexamethasone (VCD). A randomized trial comparing VTD with VCD as an induction therapy was conducted in patients with newly diagnosed MM. The overall response rate after four cycles of VCD was inferior to that after four cycles of VTD (83.4% vs. 92.3%, P=0.01). The incidence of grade 3–4 peripheral neuropathy was higher after VTD regimen than after VCD regimen (7.7% vs. 2.9%, P=0.05), but hematologic adverse events was frequent in VCD regimen [31]. In a matched-pair analysis by Cavo et al. [32], VTD, compared to VCD, showed a threefold or higher increase in CR rate in patients with newly diagnosed MM, but resulted in higher rates of grade 3 or 4 peripheral neuropathy. Therefore, VTD regimen was superior to VCD in terms of response rate, but had a problem of high incidence of peripheral neuropathy.

Substitution of lenalidomide for thalidomide in VTD regimen may decrease peripheral neuropathy and increase response rate. The phase 3 PETHEMA/GEM2012 study evaluated the efficacy and safety of six cycles of bortezomib, lenalidomide, and dexamethasone (VRD) in 458 patients aged ≤65 years with newly diagnosed MM [33]. The CR rate after VRD induction therapy was 33.4%, and the rate of VGPR or better was 66.6%. Responses deepened throughout VRD induction therapy. Furthermore, grade 3 or 4 peripheral neuropathy occurred in only 3.9% of patients. Therefore, VRD regimen is currently being considered as a standard induction therapy.

Carfilzomib and ixazomib are promising second-generation proteasome inhibitors and have been evaluated as an induction therapy with lenalidomide and dexamethasone for patients with newly diagnosed MM. In the FORTE trial, 158 patients received four cycles of carfilzomib, lenalidomide, and dexamethasone (KRd) induction followed by HDT/ASCT, and the rate of VGPR or better prior to maintenance was 89% in all patients and 86% in patients with R-ISS II and III [34]. Ixazomib, lenalidomide, and dexamethasone (IRd) regimen has been evaluated in a phase 2 study by IFM. Following three cycles of IRd, the rate of VGPR or better was 36%, with 12% CR. No development of grade 3 or 4 peripheral neuropathy was observed [35].

The addition of an agent to a three-drug regimen was expected to improve efficacy prior to ASCT. However, the result of the randomized EVOLUTION trial, which compared VRD plus cyclophosphamide (VDCR), VRD, and VCD, was disappointing. VDCR did not improve response rate, and its toxicities appeared to be higher than that of the other regimens, especially hematologic toxicity [36]. However, the development of monoclonal antibodies, such as daratumumab or elotuzumab, has revived our hope for four-drug regimens. In the CASSIOPEIA trial, which compared daratumumab plus VTD (D-VTD) with VTD for transplant-eligible patients, the addition of daratumumab resulted in deep response after four cycles of induction (the rate of VGPR or better, 64.9% vs. 56.1%) and improved the median PFS. Furthermore, discontinuation rate during induction was slightly lower in the D-VTD group than in the VTD group (4% vs. 6%), and the occurrence of serious adverse events, including infection, was not different in both groups [37]. Therefore, the addition of daratumumab in an induction therapy may be considered for patients who are expected to show inadequate response to three-drug regimens, such as VTD or VRD.

Transplant-ineligible patients

The main result of recent clinical trials in transplant-ineligible patients for newly diagnosed MM are summerized in Table 2.

Table 2 Summary of major recent clinical trials in transplant-ineligible patients with newly diagnosed multiple myeloma.

Ref.RegimenCycle of inductionNCR rateMedian PFSMedian OS
San Miguel et al. [38]VMP vs.934430%19.9 moNot reached
MP3384%13.1 mo43.1 mo
Facon et al. [40]KMP vs.947825.9%22.3 moNA
VMP47723.1%22.1 mo
Facon et al. [41]Rd continuous vs.Continuous5350.2226.0 mo59.1 mo
MPT18 vs.1854112%21.9 mo49.1 mo
Rd181854720%21.0 mo62.3 mo
O’Donnell et al. [43]VRD lite95044%35.1 moNA
Facon et al. [44]DRd vs.36847.6%Not reachedNA
Rd36924.9%31.9 months
Mateos et al. [45]D-VMP vs.935042.6%Not reachedNA
VMP35624.4%18.1 mo

Abbreviations: CR, complete response; DRd, daratumumab, lenalidomide, and dexamethasone; D-VMP, daratumumab, bortezomib, melphalan, and prednisone; MP, melphalan and prednisone; MPT, melphalan, prednisone, and thalidomide; NA, not available; OS, overall survival; PFS, progression-free survival; Rd, lenalidomide and dexamethasone; Ref, reference; VMP, bortezomib, melphalan, and prednisone; VRD, bortezomib, lenalidomide, and dexamethasone.



Generally, transplant-ineligible patients receive 8 or 12 cycles of induction therapy followed by maintenance therapy until progression. Melphalan-containing regimens, such as melphalan, prednisone, and thalidomide (MPT) regimen or bortezomib, melphalan, and prednisone (VMP) regimen, were the main induction therapies for transplant-ineligible patients. In the phase 3 VISTA trial, which compared VMP with MP, CR rate was higher in the VMP group than in the MP group (30% vs. 4%). In addition, the median PFS was significantly higher in the VMP group than in the MP group (24.0 mo vs. 16.6 mo, P<0.001). However, addition of bortezomib to MP resulted in high incidence of peripheral neuropathy and gastrointestinal toxicity [38]. In real-world practice, especially in Asian countries, VMP was also an effective induction therapy, but it caused intolerance to the original treatment schedule. Serious adverse events developed in 68.4% of patients, 14.7% of which resulted in death. Development of peripheral neuropathy was the leading cause of discontinuation [39]. Therefore, bortezomib must be injected subcutaneously to decrease peripheral neuropathy, and early dose adjustment including weekly injection of bortezomib is essential according to frailty status and adverse events. Recently, the results of the phase III CLARION study, which compared carfilzomib, melphalan, and prednisone (KMP) regimen with VMP regimen in transplant-eligible newly diagnosed MM patients, were reported [40]. The patients received nine cycles of induction therapy. Substitution of carfilzomib for bortezomib in VMP regimen did not improve outcomes in terms of CR rate, median PFS, and OS. Although grade ≥2 peripheral neuropathy was lower in the KMP group than in the VMP group (2.5% vs. 35.1%), acute renal failure and cardiac failure frequently developed in the KMP group.

Recently, non-alkylator-containing regimens have emerged as effective induction therapies for transplant-eligible patients. The FIRST trial evaluated survival outcomes in patients with transplant-ineligible newly diagnosed MM treated with continuous treatment of lenalidomide and dexamethasone (Rd), 18 cycles of MPT, and 18 cycles of Rd. Continuous Rd significantly improved median PFS, compared to MPT and fixed duration of Rd (26.0 vs. 21.9 vs. 21.0 mo, respectively). Median OS with continuous Rd was superior to that with MPT, but not to that with fixed Rd (59.1 vs. 49.1 vs. 62.3 mo, respectively) [41]. Therefore, based on the FIRST trial, Rd was considered a preferred regimen for induction in transplant-ineligible patients. In addition, Rd is used as a backbone of various induction regimens for transplant-ineligible patients. In a subgroup analysis of the SWOG-S0777 trial, compared to Rd, VRD showed significantly improved survival benefits in patients aged ≥65 years [42]. However, there was a concern regarding the dosing schedule of VRD in elderly patients. Reduced intensity of VRD (VRD lite) with lenalidomide 15 mg at days 1–21 was evaluated in 50 elderly patients in a phase II trial [43]. Thirty-seven (74%) of patients completed nine cycles of VRD lite induction therapy, whereas 4% of patients discontinued treatment owing to adverse events. The overall response rate was 86%, including 44% CR or better. Therefore, VRD was also accepted as a standard regimen for transplant-ineligible patients, and dose adjustment according to age and frailty was also needed. The interim analysis results of a randomized phase III trial (MAIA) comparing daratumumab, lenalidomide, and dexamethasone (DRd) with Rd in transplant-ineligible patients have been reported [44]. Patients received DRd or Rd until progression. The rate of CR or better was significantly higher in the DRd group than in the Rd group (47.6% vs. 24.9%, P<0.001), and the median PFS in the DRd group was significantly improved compared to that in the Rd group (not reached vs. 31.9 mo, P<0.001). Compared to Rd, infusion of daratumumab resulted in 2.7% of grade 3 or 4 infusion-related reaction and increased infection rates. Therefore, DRd induction therapy was very effective and tolerable and expected to become a standard induction therapy for transplant-ineligible patients in the near future.

A four-drug regimen was also investigated in transplant-ineligible patients with newly diagnosed MM. A combination of daratumumab, bortezomib, melphalan, and prednisone (D-VMP) was compared with VMP in a randomized phase 3 trial (ALCYONE trial) [45]. D-VMP, compared with VMP, resulted in significantly higher rate of CR after induction therapy (42.6% vs. 24.4%, P<0.001) and reduced risk of progression to 50% (HR, 0.50; 95% CI, 0.38–0.65; P<0.001). In addition, compared with VMP, D-VMP showed acceptable safety profile, but more frequent development of grade 3 or 4 infections. Therefore, four-drug regimens with daratumumab were considered promising induction therapies for transplant-eligible patients.

Three-drug regimens with proteasome inhibitors or immunomodulatory drugs are currently accepted as standard induction therapies for patients with newly diagnosed MM. Substitution of a second-generation inhibitor for bortezomib increased tolerability with high efficacy in transplant-eligible patients. Addition of monoclonal antibodies led to the availability of a four-drug regimen for transplant-eligible patients, and DRd and D-VMP are emerging as promising induction therapies for transplant-ineligible patients. To date, individualized and optimized therapies according to risk status are not possible; however, selection of an induction therapy should at least be determined with considerations of the efficacy and toxicity of the regimen and the patients’ frailty.

No potential conflicts of interest relevant to this article were reported.

  1. Kyle RA, Rajkumar SV. Multiple myeloma. N Engl J Med 2004;351:1860-73.
    Pubmed CrossRef
  2. Siegel RL, Miller KD, Jemal A. Cancer statistics, 2015. CA Cancer J Clin 2015;65:5-29.
    Pubmed CrossRef
  3. Jung KW, Won YJ, Kong HJ, et al. Cancer statistics in Korea: incidence, mortality, survival, and prevalence in 2012. Cancer Res Treat 2015;47:127-41.
    Pubmed KoreaMed CrossRef
  4. Lee JH, Lee DS, Lee JJ, et al. Multiple myeloma in Korea: past, present, and future perspectives. Experience of the Korean Multiple Myeloma Working Party. Int J Hematol 2010;92:52-7.
    Pubmed CrossRef
  5. Barlogie B, Smith L, Alexanian R. Effective treatment of advanced multiple myeloma refractory to alkylating agents. N Engl J Med 1984;310:1353-6.
    Pubmed CrossRef
  6. Child JA, Morgan GJ, Davies FE, et al. High-dose chemotherapy with hematopoietic stem-cell rescue for multiple myeloma. N Engl J Med 2003;348:1875-83.
    Pubmed CrossRef
  7. Kristinsson SY, Landgren O, Dickman PW, Derolf AR, Björkholm M. Patterns of survival in multiple myeloma: a population-based study of patients diagnosed in Sweden from 1973 to 2003. J Clin Oncol 2007;25:1993-9.
    Pubmed CrossRef
  8. Kumar SK, Rajkumar SV, Dispenzieri A, et al. Improved survival in multiple myeloma and the impact of novel therapies. Blood 2008;111:2516-20.
    Pubmed KoreaMed CrossRef
  9. 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;127:2955-62.
    Pubmed KoreaMed CrossRef
  10. Greipp PR, San Miguel J, Durie BG, et al. International staging system for multiple myeloma. J Clin Oncol 2005;23:3412-20.
    Pubmed CrossRef
  11. 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;33:2863-9.
    Pubmed KoreaMed CrossRef
  12. Hanamura I, Stewart JP, Huang Y, et al. Frequent gain of chromosome band 1q21 in plasma-cell dyscrasias detected by fluorescence in situ hybridization: incidence increases from MGUS to relapsed myeloma and is related to prognosis and disease progression following tandem stem-cell transplantation. Blood 2006;108:1724-32.
    Pubmed KoreaMed CrossRef
  13. Boyd KD, Ross FM, Chiecchio L, et al. A novel prognostic model in myeloma based on co-segregating adverse FISH lesions and the ISS: analysis of patients treated in the MRC myeloma IX trial. Leukemia 2012;26:349-55.
    Pubmed KoreaMed CrossRef
  14. Walker BA, Mavrommatis K, Wardell CP, et al. A high-risk, double-hit, group of newly diagnosed myeloma identified by genomic analysis. Leukemia 2019;33:159-70.
    Pubmed KoreaMed CrossRef
  15. Decaux O, Lode 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 Myelome. J Clin Oncol 2008;26:4798-805.
    Pubmed CrossRef
  16. 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;109:2276-84.
    Pubmed CrossRef
  17. Kuiper R, Broyl A, de Knegt Y, et al. A gene expression signature for high-risk multiple myeloma. Leukemia 2012;26:2406-13.
    Pubmed CrossRef
  18. Moulopoulos LA, Dimopoulos MA, Kastritis E, et al. Diffuse pattern of bone marrow involvement on magnetic resonance imaging is associated with high risk cytogenetics and poor outcome in newly diagnosed, symptomatic patients with multiple myeloma: a single center experience on 228 patients. Am J Hematol 2012;87:861-4.
    Pubmed CrossRef
  19. Bartel TB, Haessler J, Brown TL, et al. F18-fluorodeoxyglucose positron emission tomography in the context of other imaging techniques and prognostic factors in multiple myeloma. Blood 2009;114:2068-76.
    Pubmed KoreaMed CrossRef
  20. Usmani SZ, Mitchell A, Waheed S, et al. Prognostic implications of serial 18-fluoro-deoxyglucose emission tomography in multiple myeloma treated with total therapy 3. Blood 2013;121:1819-23.
    Pubmed KoreaMed CrossRef
  21. Jung SH, Kwon SY, Min JJ, et al. (18)F-FDG PET/CT is useful for determining survival outcomes of patients with multiple myeloma classified as stage II and III with the Revised International Staging System. Eur J Nucl Med Mol Imaging 2019;46:107-15.
    Pubmed CrossRef
  22. Attal M, Harousseau JL, Stoppa AM, et al. A prospective, randomized trial of autologous bone marrow transplantation and chemotherapy in multiple myeloma. Intergroupe Francais du Myelome. N Engl J Med 1996;335:91-7.
    Pubmed CrossRef
  23. Koreth J, Cutler CS, Djulbegovic B, et al. High-dose therapy with single autologous transplantation versus chemotherapy for newly diagnosed multiple myeloma: A systematic review and meta-analysis of randomized controlled trials. Biol Blood Marrow Transplant 2007;13:183-96.
    Pubmed CrossRef
  24. Cavo M, Hájek R, Pantani L, et al. Autologous stem cell transplantation versus bortezomib-melphalan-prednisone for newly diagnosed multiple myeloma: second interim analysis of the phase 3 EMN02/HO95 Study. Blood (ASH Annual Meeting Abstracts) 2017;130(Suppl 1):397.
  25. Dhakal B, Szabo A, Chhabra S, et al. Autologous transplantation for newly diagnosed multiple myeloma in the era of novel agent induction: a systematic review and meta-analysis. JAMA Oncol 2018;4:343-50.
    Pubmed KoreaMed CrossRef
  26. Kumar SK, Dingli D, Lacy MQ, et al. Autologous stem cell transplantation in patients of 70 years and older with multiple myeloma: Results from a matched pair analysis. Am J Hematol 2008;83:614-7.
    Pubmed CrossRef
  27. Bashir Q, Shah N, Parmar S, et al. Feasibility of autologous hematopoietic stem cell transplant in patients aged ≥70 years with multiple myeloma. Leuk Lymphoma 2012;53:118-22.
    Pubmed CrossRef
  28. Cavo M, Tacchetti P, Patriarca F, et al. Bortezomib with thalidomide plus dexamethasone compared with thalidomide plus dexamethasone as induction therapy before, and consolidation therapy after, double autologous stem-cell transplantation in newly diagnosed multiple myeloma: a randomised phase 3 study. Lancet 2010;376:2075-85.
    Pubmed CrossRef
  29. Rosiñol L, Oriol A, Teruel AI, et al. Superiority of bortezomib, thalidomide, and dexamethasone (VTD) as induction pretransplantation therapy in multiple myeloma: a randomized phase 3 PETHEMA/GEM study. Blood 2012;120:1589-96.
    Pubmed CrossRef
  30. Moreau P, Avet-Loiseau H, Facon T, et al. Bortezomib plus dexamethasone versus reduced-dose bortezomib, thalidomide plus dexamethasone as induction treatment before autologous stem cell transplantation in newly diagnosed multiple myeloma. Blood 2011;118:5752-8; quiz 5982.
    Pubmed CrossRef
  31. Moreau P, Hulin C, Macro M, et al. VTD is superior to VCD prior to intensive therapy in multiple myeloma: results of the prospective IFM2013-04 trial. Blood 2016;127:2569-74.
    Pubmed CrossRef
  32. Cavo M, Pantani L, Pezzi A, et al. Bortezomib-thalidomide-dexamethasone (VTD) is superior to bortezomib-cyclophosphamide-dexamethasone (VCD) as induction therapy prior to autologous stem cell transplantation in multiple myeloma. Leukemia 2015;29:2429-31.
    Pubmed CrossRef
  33. Rosiñol L, Oriol A, Rios R, et al. Bortezomib, lenalidomide, and dexamethasone as induction therapy prior to autologous transplant in multiple myeloma. Blood 2019;134:1337-45.
    Pubmed KoreaMed CrossRef
  34. Gay F, Cerrato C, Petrucci MT, et al. Efficacy of carfilzomib lenalidomide dexamethasone (KRd) with or without transplantation in newly diagnosed myeloma according to risk status: Results from the FORTE trial. J Clin Oncol (ASCO Annual Meeting) 2019;37(Suppl 15):abst 8002.
    CrossRef
  35. Moreau P, Hulin C, Caillot D, et al. Ixazomib-lenalidomide-dexamethasone (IRd) combination before and after autologous stem cell transplantation (ASCT) followed by ixazomib maintenance is a safe and effective strategy in patients with newly diagnosed multiple myeloma (NDMM): A phase 2 study from the Intergroupe Francophone Du MyéLome (IFM). Blood (ASH Annual Meeting Abstracts) 2017;130(Suppl 1):2021.
    CrossRef
  36. Kumar S, Flinn I, Richardson PG, et al. Randomized, multicenter, phase 2 study (EVOLUTION) of combinations of bortezomib, dexamethasone, cyclophosphamide, and lenalidomide in previously untreated multiple myeloma. Blood 2012;119:4375-82.
    Pubmed CrossRef
  37. Moreau P, Attal M, Hulin C, et al. Bortezomib, thalidomide, and dexamethasone with or without daratumumab before and after autologous stem-cell transplantation for newly diagnosed multiple myeloma (CASSIOPEIA): a randomised, open-label, phase 3 study. Lancet 2019;394:29-38.
    Pubmed CrossRef
  38. 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;359:906-17.
    Pubmed CrossRef
  39. Kim MK, Kim K, Min CK, et al. A prospective, open-label, multicenter, observational study to evaluate the efficacy and safety of bortezomib-melphalan-prednisone as initial treatment for autologous stem cell transplantation-ineligible patients with multiple myeloma. Oncotarget 2017;8:37605-18.
    Pubmed KoreaMed CrossRef
  40. Facon T, Lee JH, Moreau P, et al. Carfilzomib or bortezomib with melphalan-prednisone for transplant-ineligible patients with newly diagnosed multiple myeloma. Blood 2019;133:1953-63.
    Pubmed CrossRef
  41. Facon T, Dimopoulos MA, Dispenzieri A, et al. Final analysis of survival outcomes in the phase 3 FIRST trial of up-front treatment for multiple myeloma. Blood 2018;131:301-10.
    Pubmed KoreaMed CrossRef
  42. Durie BGM, Hoering A, Abidi MH, et al. Bortezomib with lenalidomide and dexamethasone versus lenalidomide and dexamethasone alone in patients with newly diagnosed myeloma without intent for immediate autologous stem-cell transplant (SWOG S0777): a randomised, open-label, phase 3 trial. Lancet 2017;389:519-27.
    Pubmed KoreaMed CrossRef
  43. O'Donnell EK, Laubach JP, Yee AJ, et al. A phase 2 study of modified lenalidomide, bortezomib and dexamethasone in transplant-ineligible multiple myeloma. Br J Haematol 2018;182:222-30.
    Pubmed KoreaMed CrossRef
  44. Facon T, Kumar S, Plesner T, et al. Daratumumab plus lenalidomide and dexamethasone for untreated myeloma. N Engl J Med 2019;380:2104-15.
    Pubmed CrossRef
  45. Mateos MV, Dimopoulos MA, Cavo M, et al. Daratumumab plus bortezomib, melphalan, and prednisone for untreated myeloma. N Engl J Med 2018;378:518-28.
    Pubmed CrossRef

Article

Review Article

Blood Res 2020; 55(S1): S37-S42

Published online July 31, 2020 https://doi.org/10.5045/br.2020.S007

Copyright © The Korean Society of Hematology.

Frontline therapy for newly diagnosed patients with multiple myeloma

Sung-Hoon Jung1, Jae-Cheol Jo2, Ga-Young Song1, Seo-Yeon Ahn1, Deok-Hwan Yang1, Jae-Sook Ahn1, Hyeoung-Joon Kim1, Je-Jung Lee1

1Department of Hematology-Oncology, Chonnam National University Hwasun Hospital, Hwasun, 2Department of Hematology and Oncology, Ulsan University Hospital, University of Ulsan College of Medicine, Ulsan, Korea

Correspondence to:Je-Jung Lee, M.D., Ph.D.
Department of Hematology-Oncology, Chonnam National University Hwasun Hospital, 322 Seoyangro, Hwasun 58128, Korea
E-mail: drjejung@chonnam.ac.kr

Received: November 13, 2019; Revised: January 22, 2020; Accepted: January 30, 2020

This is an Open Access article distributed unAcute myeloid leukemia, New FDA approvalsder the terms of the Creative Commons Attribution Non-Commercial License (http://creativecommons.org/licenses/by-nc/4.0) which permits unrestricted non-commercial use, distribution, and reproduction in any medium, provided the original work is properly cited.

Abstract

Since the introduction of an alkylator to the treatment of multiple myeloma (MM), new effective agents have been developed, such as immunomodulatory drugs including thalidomide, lenalidomide, and pomalidomide; proteasome inhibitors including bortezomib, carfilzomib, and ixazomib; monoclonal antibodies including daratumumab and elotuzumab; and deacetylase inhibitors including panobinostat. Numerous regimens with these new agents have been developed and they have contributed in improving survival outcomes in MM patients. In addition, the recommended therapies for newly diagnosed MM change every year based on the results of clinical trials. This review will discusses the appropriate induction therapies based on recent clinical trials for patients with newly diagnosed MM.

Keywords: Multiple myeloma, Induction therapy, Prognosis

INTRODUCTION

Multiple myeloma (MM) is a clonal B-cell malignancy characterized by anemia, hypercalcemia, renal insufficiency, osteolytic bone lesions, and decreased immune function due to aberrant proliferation of malignant plasma cells in the bone marrow [1]. MM is the second-most common hematologic malignancy and it accounts for 10% of all hematologic malignancies in the United States [2]. In Korea, the incidence of MM is lower than that in Western countries, but is increasing rapidly over the last three decades. According to the 2012 Korean data on national cancer statistics, the age-standardized incidence rate of MM in 2012 was 10 times higher than that 20 years ago [3, 4]. This increase in MM incidence in Korea is presumed to be due to aging, exposure to environmental risk and improved early detection by the expansion of health insurance, relevant education of health providers, and public awareness of this disease.

Treatment options for MM have rapidly expanded in the recent decades. In the 1980s, induction therapy with alkylating agents, such as anthracyclines and corticosteroids, as well as high-dose chemotherapy followed by autologous stem cell transplantation (HDT/ASCT) were the main treatment approaches for MM [5, 6]. However, these treatment approaches have markedly changed since the introduction of several more effective novel agents, such as proteasome inhibitors, including bortezomib, carfilzomib, and ixazomib, and immunomodulatory drugs, including thalidomide, lenalidomide, and pomalidomide. Treatment with these agents improves response rates in relapsed or refractory MM. Furthermore, their use in induction therapy resulted in considerable improvement of outcomes and extended overall survival (OS) times [7, 8]. Numerous regimens with these agents have been developed and the result of clinical trials with new regimens are reported each year. Furthermore, the recent development of monoclonal antibodies and histone deacetylase inhibitor for MM have further expanded the treatment landscape by improving deep response rate and response duration.

The purpose of this review was to suggest the optimal combination for induction therapy based on recent clinical data for newly diagnosed patients with MM.

GENERAL APPROACH FOR SELECTING INITIAL THERAPY

Risk stratification

After being diagnosed with MM, patients must be evaluated for risk classification. Although the introduction of effective new agents for MM has improved survival outcomes, response to treatment is highly variable. Some patients may survive for more than 10 years, whereas others with an aggressive course may survive for less than 2 years. In general, high-risk MM is defined as the disease subgroup with a median OS of less than 3 years in transplant-eligible patients and less than 2 years in transplant-ineligible patients [9]. Identification of this subgroup at diagnosis would be beneficial to the personalized and intensive treatment approach from the time of induction therapy.

There have been numerous studies of accurate prognostic classification, and various prognostic staging systems have been proposed. Among them, the International Staging System (ISS) proposed by Greipp et al. [10] in 2005 was a preferred staging system. The ISS was developed using survival data from 10,750 patients from 1981 to 2002. However, several data of patients treated with novel agents were included in the analysis for ISS, and there was a debate for the prognosis value of ISS in the era of novel agents. In 2015, the Revised ISS (R-ISS) was suggested by the International Myeloma Working Group to improve risk stratification by adding cytogenetic abnormalities and serum lactate dehydrogenase (LDH) to the original ISS [11]. Cytogenetic abnormalities in R-ISS included del(17p), t(4;14), and/or t(14;16). Using these parameters, the prognostic power of the R-ISS was improved compared to that of the ISS, and showed predictive value independent of patients’ age, performance of ASCT, and treatment received. In addition to the cytogenetic abnormalities included in the R-ISS, t(14;20), gains/amplification of 1q21, and del(1p) are also considered high-risk features [9, 12]. Accumulation of these cytogenetic abnormalities is associated with poor prognosis. The study of 1,069 MM patients treated in the Medical Research Council (MRC) Myeloma IX trial firstly showed the poor prognosis of patients who had had any two of the following abnormalities: 1) adverse translocations t(4;14), t(14;16), and t(14;20); 2) gain(1q); 3) del(17p) [13]. In addition, recent genome-wide analysis of the large data set of molecular and clinical data from newly diagnosed MM, ‘double hit’ myeloma was defined as the presence of ≥1 of the following two adverse genetic factors: a) bi-allelic TP53 inactivation and b) amplification (≥4 copies) of CKS1B (1Q21). It constituted 6.1% of the patients and was associated with median progression-free survival (PFS) of 15.4 months and median OS of 20.7 months despite modern therapies [14]. Analysis of molecular subtype by gene expression profiling (GEP) for MM is expected to provide additional prognostic information and to help making therapeutic decisions. Several studies have reported that 15-gene, 70-gene, or 92-gene models based on GEP had prognostic significance in MM [15-17].

Imaging at diagnosis, such as MRI and 18F-fluorodeoxyglucose (18F-FDG) positron emission tomography/computed tomography (PET/CT), has been shown to be associated with prognosis. The diffuse pattern of marrow infiltration in MRI was associated with inferior survival than that with other MRI patterns, such as focal, variegated, and normal MRI patterns [18]. In addition, the number of FDG-avid focal lesions, presence of extramedullary disease, and maximum standardized uptake value have been reported as reliable prognostic 18F-FDG PET/CT parameters in MM [19-21].

Assessment of eligibility for ASCT

The Intergroupe Francophone du Myelome (IFM) study was the first randomized trial showing the superiority of high-dose melphalan and total body irradiation followed by ASCT over the conventional chemotherapy in terms of response rate, event-free survival, and OS [22]. Subsequent randomized trials have also shown that HDT/ASCT, compare to conventional chemotherapy, improved response rates and survival [6, 23]. Although there are some debates regarding the role of HDT/ASCT in MM with the emergence of new and more effective agents, upfront ASCT did lead to better response and longer PFS in a recent randomized phase 3 EMN02/HO95 trial [HR, 0.76; 95% confidence interval (CI), 0.64–0.91; P=0.002] [24]. In addition, a meta-analysis of three large phase 3 randomized controlled trials showed the superiority of upfront ASCT over novel agents [25]. Therefore, assessment of eligibility for ASCT at diagnosis is important to decide treatment strategy for MM. Transplant-eligible patients typically receive four to six cycles of induction therapy, followed by HDT/ASCT and then maintenance therapy until progression.

Age, generally age of 65 years, was considered a main factor in determining ASCT. However, recently, HDT/ASCT has been extended to older patients and has shown comparable outcome to that in younger patients [26, 27]. It is necessary to determine whether ASCT will proceed by evaluating frailty parameters, including performance status, organ function, and comorbidities, rather than age alone.

OPTIMAL INDUCTION THERAPY

Transplant-eligible patients

The main result of recent clinical trials in transplant-eligible patients with newly diagnosed MM are summarized in Table 1. A combination regimen of three drugs including a proteasome inhibitor is preferred for induction therapy in MM. Bortezomib, thalidomide, and dexamethasone (VTD) regimen is a preferred regimen and has been evaluated in three randomized phase III trials [28-30]. Although the dose intensity and treatment cycle of VTD were different in three randomized trials, six cycles of VTD prior to ASCT showed superior CR rate than that shown by three cycles of VTD (35% vs. 19%). In the IFM trial, patients received four cycles of VTD with lower bortezomib dose of 1.0 mg/m2 and CR rates prior to ASCT of only 13%. However, the incidence of grade 3 or 4 peripheral neuropathy was lower with reduced VTD than with four cycles of VTD or six cycles of VTD (3% vs. 10% vs. 14%). Therefore, the dose intensity and treatment duration of VTD regimen were important to achieve deep response, but increased dosing and treatment cycle were also associated with the occurrence of peripheral neuropathy.

Table 1 . Summary of major recent clinical trials in transplant-eligible patients with newly diagnosed multiple myeloma..

Ref.RegimenCycle of inductionNCR rate prior to transplantMedian PFSMedian OS
Cavo et al. [28]VTD vs.323619%3-year PFS; 68%3-year OS; 86%
TD2385%vs. 56%vs. 84%
Rosiñol et al. [29]VTD vs.613035%56.2 mo4-year OS; 74%
TD vs.12714%28.2 movs. 70%
VBMCP/VBAD/B1290.2135.3 movs. 65%
Moreau et al. [31]VTD vs.416913%NANA
VCD1698.9%
Rosiñol et al. [33]VRD645833.4%NANA
Moreau et al. [37]D-VTD vs.454339%18 mo PFS; 93%NA
VTD54226%vs. 85%

Abbreviations: CR, complete response; D-VTD; daratumumab, bortezomib, thalidomide, and dexamethasone; NA, not evaluable; OS, overall survival; PFS, progression-free survival; Ref, reference; TD, thalidomide and dexamethasone; VBMCP/VBAD, vincristine, VCD, bortezomib, cyclophosphamide, and dexamethasone; VRD, bortezomib, lenalidomide, and dexamethasone; VTD, bortezomib, thalidomide, and dexamethasone..



Another effective regimen was bortezomib, cyclophosphamide, and dexamethasone (VCD). A randomized trial comparing VTD with VCD as an induction therapy was conducted in patients with newly diagnosed MM. The overall response rate after four cycles of VCD was inferior to that after four cycles of VTD (83.4% vs. 92.3%, P=0.01). The incidence of grade 3–4 peripheral neuropathy was higher after VTD regimen than after VCD regimen (7.7% vs. 2.9%, P=0.05), but hematologic adverse events was frequent in VCD regimen [31]. In a matched-pair analysis by Cavo et al. [32], VTD, compared to VCD, showed a threefold or higher increase in CR rate in patients with newly diagnosed MM, but resulted in higher rates of grade 3 or 4 peripheral neuropathy. Therefore, VTD regimen was superior to VCD in terms of response rate, but had a problem of high incidence of peripheral neuropathy.

Substitution of lenalidomide for thalidomide in VTD regimen may decrease peripheral neuropathy and increase response rate. The phase 3 PETHEMA/GEM2012 study evaluated the efficacy and safety of six cycles of bortezomib, lenalidomide, and dexamethasone (VRD) in 458 patients aged ≤65 years with newly diagnosed MM [33]. The CR rate after VRD induction therapy was 33.4%, and the rate of VGPR or better was 66.6%. Responses deepened throughout VRD induction therapy. Furthermore, grade 3 or 4 peripheral neuropathy occurred in only 3.9% of patients. Therefore, VRD regimen is currently being considered as a standard induction therapy.

Carfilzomib and ixazomib are promising second-generation proteasome inhibitors and have been evaluated as an induction therapy with lenalidomide and dexamethasone for patients with newly diagnosed MM. In the FORTE trial, 158 patients received four cycles of carfilzomib, lenalidomide, and dexamethasone (KRd) induction followed by HDT/ASCT, and the rate of VGPR or better prior to maintenance was 89% in all patients and 86% in patients with R-ISS II and III [34]. Ixazomib, lenalidomide, and dexamethasone (IRd) regimen has been evaluated in a phase 2 study by IFM. Following three cycles of IRd, the rate of VGPR or better was 36%, with 12% CR. No development of grade 3 or 4 peripheral neuropathy was observed [35].

The addition of an agent to a three-drug regimen was expected to improve efficacy prior to ASCT. However, the result of the randomized EVOLUTION trial, which compared VRD plus cyclophosphamide (VDCR), VRD, and VCD, was disappointing. VDCR did not improve response rate, and its toxicities appeared to be higher than that of the other regimens, especially hematologic toxicity [36]. However, the development of monoclonal antibodies, such as daratumumab or elotuzumab, has revived our hope for four-drug regimens. In the CASSIOPEIA trial, which compared daratumumab plus VTD (D-VTD) with VTD for transplant-eligible patients, the addition of daratumumab resulted in deep response after four cycles of induction (the rate of VGPR or better, 64.9% vs. 56.1%) and improved the median PFS. Furthermore, discontinuation rate during induction was slightly lower in the D-VTD group than in the VTD group (4% vs. 6%), and the occurrence of serious adverse events, including infection, was not different in both groups [37]. Therefore, the addition of daratumumab in an induction therapy may be considered for patients who are expected to show inadequate response to three-drug regimens, such as VTD or VRD.

Transplant-ineligible patients

The main result of recent clinical trials in transplant-ineligible patients for newly diagnosed MM are summerized in Table 2.

Table 2 . Summary of major recent clinical trials in transplant-ineligible patients with newly diagnosed multiple myeloma..

Ref.RegimenCycle of inductionNCR rateMedian PFSMedian OS
San Miguel et al. [38]VMP vs.934430%19.9 moNot reached
MP3384%13.1 mo43.1 mo
Facon et al. [40]KMP vs.947825.9%22.3 moNA
VMP47723.1%22.1 mo
Facon et al. [41]Rd continuous vs.Continuous5350.2226.0 mo59.1 mo
MPT18 vs.1854112%21.9 mo49.1 mo
Rd181854720%21.0 mo62.3 mo
O’Donnell et al. [43]VRD lite95044%35.1 moNA
Facon et al. [44]DRd vs.36847.6%Not reachedNA
Rd36924.9%31.9 months
Mateos et al. [45]D-VMP vs.935042.6%Not reachedNA
VMP35624.4%18.1 mo

Abbreviations: CR, complete response; DRd, daratumumab, lenalidomide, and dexamethasone; D-VMP, daratumumab, bortezomib, melphalan, and prednisone; MP, melphalan and prednisone; MPT, melphalan, prednisone, and thalidomide; NA, not available; OS, overall survival; PFS, progression-free survival; Rd, lenalidomide and dexamethasone; Ref, reference; VMP, bortezomib, melphalan, and prednisone; VRD, bortezomib, lenalidomide, and dexamethasone..



Generally, transplant-ineligible patients receive 8 or 12 cycles of induction therapy followed by maintenance therapy until progression. Melphalan-containing regimens, such as melphalan, prednisone, and thalidomide (MPT) regimen or bortezomib, melphalan, and prednisone (VMP) regimen, were the main induction therapies for transplant-ineligible patients. In the phase 3 VISTA trial, which compared VMP with MP, CR rate was higher in the VMP group than in the MP group (30% vs. 4%). In addition, the median PFS was significantly higher in the VMP group than in the MP group (24.0 mo vs. 16.6 mo, P<0.001). However, addition of bortezomib to MP resulted in high incidence of peripheral neuropathy and gastrointestinal toxicity [38]. In real-world practice, especially in Asian countries, VMP was also an effective induction therapy, but it caused intolerance to the original treatment schedule. Serious adverse events developed in 68.4% of patients, 14.7% of which resulted in death. Development of peripheral neuropathy was the leading cause of discontinuation [39]. Therefore, bortezomib must be injected subcutaneously to decrease peripheral neuropathy, and early dose adjustment including weekly injection of bortezomib is essential according to frailty status and adverse events. Recently, the results of the phase III CLARION study, which compared carfilzomib, melphalan, and prednisone (KMP) regimen with VMP regimen in transplant-eligible newly diagnosed MM patients, were reported [40]. The patients received nine cycles of induction therapy. Substitution of carfilzomib for bortezomib in VMP regimen did not improve outcomes in terms of CR rate, median PFS, and OS. Although grade ≥2 peripheral neuropathy was lower in the KMP group than in the VMP group (2.5% vs. 35.1%), acute renal failure and cardiac failure frequently developed in the KMP group.

Recently, non-alkylator-containing regimens have emerged as effective induction therapies for transplant-eligible patients. The FIRST trial evaluated survival outcomes in patients with transplant-ineligible newly diagnosed MM treated with continuous treatment of lenalidomide and dexamethasone (Rd), 18 cycles of MPT, and 18 cycles of Rd. Continuous Rd significantly improved median PFS, compared to MPT and fixed duration of Rd (26.0 vs. 21.9 vs. 21.0 mo, respectively). Median OS with continuous Rd was superior to that with MPT, but not to that with fixed Rd (59.1 vs. 49.1 vs. 62.3 mo, respectively) [41]. Therefore, based on the FIRST trial, Rd was considered a preferred regimen for induction in transplant-ineligible patients. In addition, Rd is used as a backbone of various induction regimens for transplant-ineligible patients. In a subgroup analysis of the SWOG-S0777 trial, compared to Rd, VRD showed significantly improved survival benefits in patients aged ≥65 years [42]. However, there was a concern regarding the dosing schedule of VRD in elderly patients. Reduced intensity of VRD (VRD lite) with lenalidomide 15 mg at days 1–21 was evaluated in 50 elderly patients in a phase II trial [43]. Thirty-seven (74%) of patients completed nine cycles of VRD lite induction therapy, whereas 4% of patients discontinued treatment owing to adverse events. The overall response rate was 86%, including 44% CR or better. Therefore, VRD was also accepted as a standard regimen for transplant-ineligible patients, and dose adjustment according to age and frailty was also needed. The interim analysis results of a randomized phase III trial (MAIA) comparing daratumumab, lenalidomide, and dexamethasone (DRd) with Rd in transplant-ineligible patients have been reported [44]. Patients received DRd or Rd until progression. The rate of CR or better was significantly higher in the DRd group than in the Rd group (47.6% vs. 24.9%, P<0.001), and the median PFS in the DRd group was significantly improved compared to that in the Rd group (not reached vs. 31.9 mo, P<0.001). Compared to Rd, infusion of daratumumab resulted in 2.7% of grade 3 or 4 infusion-related reaction and increased infection rates. Therefore, DRd induction therapy was very effective and tolerable and expected to become a standard induction therapy for transplant-ineligible patients in the near future.

A four-drug regimen was also investigated in transplant-ineligible patients with newly diagnosed MM. A combination of daratumumab, bortezomib, melphalan, and prednisone (D-VMP) was compared with VMP in a randomized phase 3 trial (ALCYONE trial) [45]. D-VMP, compared with VMP, resulted in significantly higher rate of CR after induction therapy (42.6% vs. 24.4%, P<0.001) and reduced risk of progression to 50% (HR, 0.50; 95% CI, 0.38–0.65; P<0.001). In addition, compared with VMP, D-VMP showed acceptable safety profile, but more frequent development of grade 3 or 4 infections. Therefore, four-drug regimens with daratumumab were considered promising induction therapies for transplant-eligible patients.

CONCLUSION

Three-drug regimens with proteasome inhibitors or immunomodulatory drugs are currently accepted as standard induction therapies for patients with newly diagnosed MM. Substitution of a second-generation inhibitor for bortezomib increased tolerability with high efficacy in transplant-eligible patients. Addition of monoclonal antibodies led to the availability of a four-drug regimen for transplant-eligible patients, and DRd and D-VMP are emerging as promising induction therapies for transplant-ineligible patients. To date, individualized and optimized therapies according to risk status are not possible; however, selection of an induction therapy should at least be determined with considerations of the efficacy and toxicity of the regimen and the patients’ frailty.

Authors’ Disclosures of Potential Conflicts of Interest

No potential conflicts of interest relevant to this article were reported.

Table 1 . Summary of major recent clinical trials in transplant-eligible patients with newly diagnosed multiple myeloma..

Ref.RegimenCycle of inductionNCR rate prior to transplantMedian PFSMedian OS
Cavo et al. [28]VTD vs.323619%3-year PFS; 68%3-year OS; 86%
TD2385%vs. 56%vs. 84%
Rosiñol et al. [29]VTD vs.613035%56.2 mo4-year OS; 74%
TD vs.12714%28.2 movs. 70%
VBMCP/VBAD/B1290.2135.3 movs. 65%
Moreau et al. [31]VTD vs.416913%NANA
VCD1698.9%
Rosiñol et al. [33]VRD645833.4%NANA
Moreau et al. [37]D-VTD vs.454339%18 mo PFS; 93%NA
VTD54226%vs. 85%

Abbreviations: CR, complete response; D-VTD; daratumumab, bortezomib, thalidomide, and dexamethasone; NA, not evaluable; OS, overall survival; PFS, progression-free survival; Ref, reference; TD, thalidomide and dexamethasone; VBMCP/VBAD, vincristine, VCD, bortezomib, cyclophosphamide, and dexamethasone; VRD, bortezomib, lenalidomide, and dexamethasone; VTD, bortezomib, thalidomide, and dexamethasone..


Table 2 . Summary of major recent clinical trials in transplant-ineligible patients with newly diagnosed multiple myeloma..

Ref.RegimenCycle of inductionNCR rateMedian PFSMedian OS
San Miguel et al. [38]VMP vs.934430%19.9 moNot reached
MP3384%13.1 mo43.1 mo
Facon et al. [40]KMP vs.947825.9%22.3 moNA
VMP47723.1%22.1 mo
Facon et al. [41]Rd continuous vs.Continuous5350.2226.0 mo59.1 mo
MPT18 vs.1854112%21.9 mo49.1 mo
Rd181854720%21.0 mo62.3 mo
O’Donnell et al. [43]VRD lite95044%35.1 moNA
Facon et al. [44]DRd vs.36847.6%Not reachedNA
Rd36924.9%31.9 months
Mateos et al. [45]D-VMP vs.935042.6%Not reachedNA
VMP35624.4%18.1 mo

Abbreviations: CR, complete response; DRd, daratumumab, lenalidomide, and dexamethasone; D-VMP, daratumumab, bortezomib, melphalan, and prednisone; MP, melphalan and prednisone; MPT, melphalan, prednisone, and thalidomide; NA, not available; OS, overall survival; PFS, progression-free survival; Rd, lenalidomide and dexamethasone; Ref, reference; VMP, bortezomib, melphalan, and prednisone; VRD, bortezomib, lenalidomide, and dexamethasone..


References

  1. Kyle RA, Rajkumar SV. Multiple myeloma. N Engl J Med 2004;351:1860-73.
    Pubmed CrossRef
  2. Siegel RL, Miller KD, Jemal A. Cancer statistics, 2015. CA Cancer J Clin 2015;65:5-29.
    Pubmed CrossRef
  3. Jung KW, Won YJ, Kong HJ, et al. Cancer statistics in Korea: incidence, mortality, survival, and prevalence in 2012. Cancer Res Treat 2015;47:127-41.
    Pubmed KoreaMed CrossRef
  4. Lee JH, Lee DS, Lee JJ, et al. Multiple myeloma in Korea: past, present, and future perspectives. Experience of the Korean Multiple Myeloma Working Party. Int J Hematol 2010;92:52-7.
    Pubmed CrossRef
  5. Barlogie B, Smith L, Alexanian R. Effective treatment of advanced multiple myeloma refractory to alkylating agents. N Engl J Med 1984;310:1353-6.
    Pubmed CrossRef
  6. Child JA, Morgan GJ, Davies FE, et al. High-dose chemotherapy with hematopoietic stem-cell rescue for multiple myeloma. N Engl J Med 2003;348:1875-83.
    Pubmed CrossRef
  7. Kristinsson SY, Landgren O, Dickman PW, Derolf AR, Björkholm M. Patterns of survival in multiple myeloma: a population-based study of patients diagnosed in Sweden from 1973 to 2003. J Clin Oncol 2007;25:1993-9.
    Pubmed CrossRef
  8. Kumar SK, Rajkumar SV, Dispenzieri A, et al. Improved survival in multiple myeloma and the impact of novel therapies. Blood 2008;111:2516-20.
    Pubmed KoreaMed CrossRef
  9. 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;127:2955-62.
    Pubmed KoreaMed CrossRef
  10. Greipp PR, San Miguel J, Durie BG, et al. International staging system for multiple myeloma. J Clin Oncol 2005;23:3412-20.
    Pubmed CrossRef
  11. 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;33:2863-9.
    Pubmed KoreaMed CrossRef
  12. Hanamura I, Stewart JP, Huang Y, et al. Frequent gain of chromosome band 1q21 in plasma-cell dyscrasias detected by fluorescence in situ hybridization: incidence increases from MGUS to relapsed myeloma and is related to prognosis and disease progression following tandem stem-cell transplantation. Blood 2006;108:1724-32.
    Pubmed KoreaMed CrossRef
  13. Boyd KD, Ross FM, Chiecchio L, et al. A novel prognostic model in myeloma based on co-segregating adverse FISH lesions and the ISS: analysis of patients treated in the MRC myeloma IX trial. Leukemia 2012;26:349-55.
    Pubmed KoreaMed CrossRef
  14. Walker BA, Mavrommatis K, Wardell CP, et al. A high-risk, double-hit, group of newly diagnosed myeloma identified by genomic analysis. Leukemia 2019;33:159-70.
    Pubmed KoreaMed CrossRef
  15. Decaux O, Lode 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 Myelome. J Clin Oncol 2008;26:4798-805.
    Pubmed CrossRef
  16. 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;109:2276-84.
    Pubmed CrossRef
  17. Kuiper R, Broyl A, de Knegt Y, et al. A gene expression signature for high-risk multiple myeloma. Leukemia 2012;26:2406-13.
    Pubmed CrossRef
  18. Moulopoulos LA, Dimopoulos MA, Kastritis E, et al. Diffuse pattern of bone marrow involvement on magnetic resonance imaging is associated with high risk cytogenetics and poor outcome in newly diagnosed, symptomatic patients with multiple myeloma: a single center experience on 228 patients. Am J Hematol 2012;87:861-4.
    Pubmed CrossRef
  19. Bartel TB, Haessler J, Brown TL, et al. F18-fluorodeoxyglucose positron emission tomography in the context of other imaging techniques and prognostic factors in multiple myeloma. Blood 2009;114:2068-76.
    Pubmed KoreaMed CrossRef
  20. Usmani SZ, Mitchell A, Waheed S, et al. Prognostic implications of serial 18-fluoro-deoxyglucose emission tomography in multiple myeloma treated with total therapy 3. Blood 2013;121:1819-23.
    Pubmed KoreaMed CrossRef
  21. Jung SH, Kwon SY, Min JJ, et al. (18)F-FDG PET/CT is useful for determining survival outcomes of patients with multiple myeloma classified as stage II and III with the Revised International Staging System. Eur J Nucl Med Mol Imaging 2019;46:107-15.
    Pubmed CrossRef
  22. Attal M, Harousseau JL, Stoppa AM, et al. A prospective, randomized trial of autologous bone marrow transplantation and chemotherapy in multiple myeloma. Intergroupe Francais du Myelome. N Engl J Med 1996;335:91-7.
    Pubmed CrossRef
  23. Koreth J, Cutler CS, Djulbegovic B, et al. High-dose therapy with single autologous transplantation versus chemotherapy for newly diagnosed multiple myeloma: A systematic review and meta-analysis of randomized controlled trials. Biol Blood Marrow Transplant 2007;13:183-96.
    Pubmed CrossRef
  24. Cavo M, Hájek R, Pantani L, et al. Autologous stem cell transplantation versus bortezomib-melphalan-prednisone for newly diagnosed multiple myeloma: second interim analysis of the phase 3 EMN02/HO95 Study. Blood (ASH Annual Meeting Abstracts) 2017;130(Suppl 1):397.
  25. Dhakal B, Szabo A, Chhabra S, et al. Autologous transplantation for newly diagnosed multiple myeloma in the era of novel agent induction: a systematic review and meta-analysis. JAMA Oncol 2018;4:343-50.
    Pubmed KoreaMed CrossRef
  26. Kumar SK, Dingli D, Lacy MQ, et al. Autologous stem cell transplantation in patients of 70 years and older with multiple myeloma: Results from a matched pair analysis. Am J Hematol 2008;83:614-7.
    Pubmed CrossRef
  27. Bashir Q, Shah N, Parmar S, et al. Feasibility of autologous hematopoietic stem cell transplant in patients aged ≥70 years with multiple myeloma. Leuk Lymphoma 2012;53:118-22.
    Pubmed CrossRef
  28. Cavo M, Tacchetti P, Patriarca F, et al. Bortezomib with thalidomide plus dexamethasone compared with thalidomide plus dexamethasone as induction therapy before, and consolidation therapy after, double autologous stem-cell transplantation in newly diagnosed multiple myeloma: a randomised phase 3 study. Lancet 2010;376:2075-85.
    Pubmed CrossRef
  29. Rosiñol L, Oriol A, Teruel AI, et al. Superiority of bortezomib, thalidomide, and dexamethasone (VTD) as induction pretransplantation therapy in multiple myeloma: a randomized phase 3 PETHEMA/GEM study. Blood 2012;120:1589-96.
    Pubmed CrossRef
  30. Moreau P, Avet-Loiseau H, Facon T, et al. Bortezomib plus dexamethasone versus reduced-dose bortezomib, thalidomide plus dexamethasone as induction treatment before autologous stem cell transplantation in newly diagnosed multiple myeloma. Blood 2011;118:5752-8; quiz 5982.
    Pubmed CrossRef
  31. Moreau P, Hulin C, Macro M, et al. VTD is superior to VCD prior to intensive therapy in multiple myeloma: results of the prospective IFM2013-04 trial. Blood 2016;127:2569-74.
    Pubmed CrossRef
  32. Cavo M, Pantani L, Pezzi A, et al. Bortezomib-thalidomide-dexamethasone (VTD) is superior to bortezomib-cyclophosphamide-dexamethasone (VCD) as induction therapy prior to autologous stem cell transplantation in multiple myeloma. Leukemia 2015;29:2429-31.
    Pubmed CrossRef
  33. Rosiñol L, Oriol A, Rios R, et al. Bortezomib, lenalidomide, and dexamethasone as induction therapy prior to autologous transplant in multiple myeloma. Blood 2019;134:1337-45.
    Pubmed KoreaMed CrossRef
  34. Gay F, Cerrato C, Petrucci MT, et al. Efficacy of carfilzomib lenalidomide dexamethasone (KRd) with or without transplantation in newly diagnosed myeloma according to risk status: Results from the FORTE trial. J Clin Oncol (ASCO Annual Meeting) 2019;37(Suppl 15):abst 8002.
    CrossRef
  35. Moreau P, Hulin C, Caillot D, et al. Ixazomib-lenalidomide-dexamethasone (IRd) combination before and after autologous stem cell transplantation (ASCT) followed by ixazomib maintenance is a safe and effective strategy in patients with newly diagnosed multiple myeloma (NDMM): A phase 2 study from the Intergroupe Francophone Du MyéLome (IFM). Blood (ASH Annual Meeting Abstracts) 2017;130(Suppl 1):2021.
    CrossRef
  36. Kumar S, Flinn I, Richardson PG, et al. Randomized, multicenter, phase 2 study (EVOLUTION) of combinations of bortezomib, dexamethasone, cyclophosphamide, and lenalidomide in previously untreated multiple myeloma. Blood 2012;119:4375-82.
    Pubmed CrossRef
  37. Moreau P, Attal M, Hulin C, et al. Bortezomib, thalidomide, and dexamethasone with or without daratumumab before and after autologous stem-cell transplantation for newly diagnosed multiple myeloma (CASSIOPEIA): a randomised, open-label, phase 3 study. Lancet 2019;394:29-38.
    Pubmed CrossRef
  38. 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;359:906-17.
    Pubmed CrossRef
  39. Kim MK, Kim K, Min CK, et al. A prospective, open-label, multicenter, observational study to evaluate the efficacy and safety of bortezomib-melphalan-prednisone as initial treatment for autologous stem cell transplantation-ineligible patients with multiple myeloma. Oncotarget 2017;8:37605-18.
    Pubmed KoreaMed CrossRef
  40. Facon T, Lee JH, Moreau P, et al. Carfilzomib or bortezomib with melphalan-prednisone for transplant-ineligible patients with newly diagnosed multiple myeloma. Blood 2019;133:1953-63.
    Pubmed CrossRef
  41. Facon T, Dimopoulos MA, Dispenzieri A, et al. Final analysis of survival outcomes in the phase 3 FIRST trial of up-front treatment for multiple myeloma. Blood 2018;131:301-10.
    Pubmed KoreaMed CrossRef
  42. Durie BGM, Hoering A, Abidi MH, et al. Bortezomib with lenalidomide and dexamethasone versus lenalidomide and dexamethasone alone in patients with newly diagnosed myeloma without intent for immediate autologous stem-cell transplant (SWOG S0777): a randomised, open-label, phase 3 trial. Lancet 2017;389:519-27.
    Pubmed KoreaMed CrossRef
  43. O'Donnell EK, Laubach JP, Yee AJ, et al. A phase 2 study of modified lenalidomide, bortezomib and dexamethasone in transplant-ineligible multiple myeloma. Br J Haematol 2018;182:222-30.
    Pubmed KoreaMed CrossRef
  44. Facon T, Kumar S, Plesner T, et al. Daratumumab plus lenalidomide and dexamethasone for untreated myeloma. N Engl J Med 2019;380:2104-15.
    Pubmed CrossRef
  45. Mateos MV, Dimopoulos MA, Cavo M, et al. Daratumumab plus bortezomib, melphalan, and prednisone for untreated myeloma. N Engl J Med 2018;378:518-28.
    Pubmed CrossRef
Blood Res
Volume 59 2024

Stats or Metrics

Share this article on

  • line

Related articles in BR

Blood Research

pISSN 2287-979X
eISSN 2288-0011
qr-code Download