Open access peer-reviewed chapter

Treatment of Patients with Newly-Diagnosed Multiple Myeloma

Written By

Ali Zahit Bolaman and Atakan Turgutkaya

Submitted: 27 May 2022 Reviewed: 08 June 2022 Published: 02 July 2022

DOI: 10.5772/intechopen.105774

From the Edited Volume

Recent Updates on Multiple Myeloma

Edited by Khalid Ahmed Al-Anazi

Chapter metrics overview

95 Chapter Downloads

View Full Metrics

Abstract

Multiple Myeloma is an incurable disease. It is responsible for 1.8% of all cancers. The median age is 69–71 years. The treatment of MM is challenging and is affected by several factors such as the patient’s age, comorbidity index, and fitness. The main combination regimen consists of the addition of proteasome inhibitors and IMIDs to steroids. In all studies conducted to date, the results obtained in transplanted patients are better than in patients who did not proceed into transplantation. Before starting treatment, risk stratification should be performed for all patients, and they should be treated accordingly. Recently, there have been advances in the treatment with the introduction of new agents, particularly monoclonal antibodies.

Keywords

  • multiple myeloma
  • cytogenetic abnormality
  • geriatric assessment
  • risk stratification
  • consolidation

1. Introduction

Multiple myeloma (MM) is characterized by clonal malignant plasma cell increase in the bone marrow. Clinical manifestations are anemia, low back pain, and infections. Hypogamoglobulinemia, osteolytic bone disease, hypercalcemia, and renal dysfunction are common in symptomatic patients. MM is responsible for 10% and 1.8% of hematologic and all malignancies, respectively. The median age for the disease is 69 and it is rare under the age of 45 [1]. The most frequent morbidity cause is bone disease due to osteolysis. It can be detected by using fluoro-deoxyglucose and (FDG) positron emission tomography/computed tomographic scans (PET/CT), whole-body computed tomography (WB-CT), or magnetic resonance imaging (MRI). PET/CT may offer anatomical and metabolic information with a sensitivity of approximately 80–90% and a specificity of 80–100% [2].

In the 1980s, MM could only be treated with alkylating agents and steroids. Later, in the 1990s, the availability of autologous stem cell transplantation (ASCT) improved the course of the disease. In the 2000s, advances were made with the first immunomodulatory (IMID’s) agent thalidomide, In time, the new generation of IMIDs (lenalidomide and pomalidomide) with fewer adverse effects, proteasome inhibitors (bortezomib, carfilzomib, and ixazomib), monoclonal antibodies and histone deacetylase inhibitors have positively impacted the survival of MM patients.

Stratification of the patients is essential for the appropriate management of newly diagnosed multiple myeloma (NDMM). Treatment can be performed according to the following subjects:

  1. Treatment of smoldering MM (controversial).

  2. Treatment for transplantation-eligible (TE) patients.

  3. Treatment for transplantation-ineligible (TI) patients.

  4. Treatment of fragile patients.

Advertisement

2. Risk stratification

The survival of MM patients varies: Some patients demonstrate more than 10 years of survival, while some patients have a limited lifespan of 2–3 years. The main reason for this is the patient’s comorbidities and the biology of the disease. Age is an important factor in treatment selection. However, there is still debate about which patient should be considered elderly [3]. The majority of authors believe that age is not the only determinant for treatment. Today, it is considered more decisive whether the patients are fit or not in the choice of treatment method. For this purpose, the Eastern Cooperative Oncology Group (ECOG) performance status can be used as a useful guide. Patients with ECOG performance status 0–1 are candidates for ASCT. The presence of comorbidities can optimally be determined by the Charlson comorbidity index. Chromosomal abnormalities demonstrated by the fluorescent in situ hybridization are also important for risk stratification which should be performed as soon as MM diagnosis is made. Table 1 demonstrates the association of cytogenetic abnormalities with prognosis, survey, and treatment.

Cytogenetic abnormalityRiskPrognosisSurvey (years)Treatment relation
All types of trisomiesStandardFavorable7–10Good response to lenalidomide
t(11;14) (q13;q32)StandardFavorable7–10
t(6;14) (p21;q32)StandardFavorable7–10
t(4;14) (p16;q32)HighAdverse5ASCT recommended
t(14;16) (q32;q23)HighAdverse525% risk of renal failure
t(14;20) (q32;q11)HighAdverse5ASCT recommended
Gain of 1q21HighAdverse5ASCT recommended
Del 17 pHighAdverse5ASCT recommended
Trisomy+ del 14UnknownUnknownUnknownNegativity due to del 17p, 14. chromosome translocations can disappear
NoneLowGood7–10Indicate to low tumor burden

Table 1.

The association of cytogenetic abnormalities with prognosis, survey, and treatment.

Treatment of patients with a high Revised-International Scoring System (R-ISS) requires a more aggressive approach. Proteosome inhibitors are especially effective in patients with a high-risk cytogenetic risk. Patients with renal involvement may also benefit from bortezomib treatment. Other factors that are effective in determining the treatment algorithm are the patient’s life expectancy, treatment preference, and the presence of extramedullary disease. Geriatric assessment is crucial for frail patients and consists of age, the activity of daily living (ADL), the Charlson Comorbidity Index (CCI), and instrumental activity of daily living (IADL) (Table 2) [4].

Age> 8076–80
Plus at least 1 of the following
ADL ≤ 4
IADL ≤4
CCI ≥ 2
≤ 75
Plus at least 2 of the following
ADL ≤ 4
IADL ≤4
CCI ≥ 2

Table 2.

Geriatric assessment index for frail patients with MM.

Advertisement

3. Treatment of transplant-eligible patients

Therapy with high-dose melphalan and ASCT is very effective in patients with MM. Intergroupe Francophone du Myelome (IFM) and EMN/H095 studies have shown that bortezomib, which is used in the induction regimen, is a beneficial drug for TE patients. TE patients were treated with at least 4 cycles of chemotherapy. Thereafter, the patients were evaluated for response. PFS with ASCT was found better than the bortezomib-melphalan-dexamethasone group (567.7 vs. 41.9 months, p = 0.0001) [5, 6].

Many centers perform transplantation when patients achieve a very good partial remission (VGPR). If patients have not achieved VGPR, two more cycles of chemotherapy can be given. The best induction regimen includes a proteasome inhibitor plus IMID plus dexamethasone. Moreau et al. compared Bortezomib, cyclophosphamide, and dexamethasone (VCD) versus bortezomib, thalidomide, and dexamethasone (VTD) combinations in induction. The overall response rate (ORR) with VTD was detected higher than VCD (92.3% vs. 83.4%, P = 0.01) [7]. Peripheral neuropathy is higher with thalidomide treatment. Lenalidomide was used instead of thalidomide in PETHEMA/GEM2012 study. VGPR or better rate was higher with VRD regimen, but neuropathy rate was lower (3.9%) [8]. Endurance Study compared VRD with carfilzomib-lenalidomide and dexamethasone regimen (CRD). ORR was similar to VRD and CRD regimens [9]. The addition of monoclonal antibodies to VTD (Cassisopea and Griffin Studies) or VRD regimen can improve transplantation results [10, 11]. Improved results with daratumumab are correlated with minimal residual disease negativity rate. These results suggest that Dara-VRD is the best regimen for induction treatment. Other effective regimens are Dara-VTD, VRD, VTD, and VCD, respectively.

The standard conditioning regimen for ASCT includes melphalan 200 mg/m2. Another drug addition to melphalan such as busulfan or bortezomib has not been found beneficial. In patients with renal dysfunction or failure, the dose of melphalan can be adjusted according to creatinine clearance [12]. The aforementioned induction regimens are summarized in Table 3.

StudyRegimenn≥ VGPR
%
≥ CR
%
PFS
(m)
OS (m)Author
IFM 2009VRD+ SCT
VRD
350
350
88 vs. 7859
47
50
36
81
82
Attal, N Eng J Med 2017
IFM 2013–2014VTD vs. VCD169
169
66 vs. 5613
8.9
Moreau, Blood 2016
PETHEMA 2012VRD4586633Rosinol, Blood 2019
GRIFFINDara-VRD104
103
90 vs. 73
22 months
42
32
Wooererhes, Blood 2020
CASSIOPEIADara-VTD
VTD
543
542
83
78
39
26
Moreau, Lancet 2019
ENDURANCEKRD
VRD
527
526
74
65
18
15
34.6
34.4
Kumar, Lancet Oncology 2020

Table 3.

Treatment regimens for TE patients.

Advertisement

4. Consolidation treatment after ASCT

Is consolidation treatment necessary after ASCT in patients with NDMM? Straka et al. evaluated the impact of bortezomib consolidation following ASCT in patients aged between 61 and 75 [13]. Consolidation treatment consisted of 4 cycles bortezomib (1.6 mg/m2 IV on days 1, 8, 15, 22) or observation only. Median PFS with bortezomib consolidation was 33.6 months while it was 29.0 months in the observation arm. They showed that consolidation treatment is useful for PFS in older patients because they received less intensive induction treatment. The generally accepted opinion today is that the agents used in the induction regimen should be given 2–4 more times post-ASCT. A randomized phase 3 study indicates that bortezomib-thalidomide-dexamethasone (VTD) is superior to thalidomide-dexamethasone (TD) as consolidation therapy after ASCT. After consolidation, the CR rate was 60.6 months in VTD while it was 46.6% months in the TD arm. Ultimately, VTD was found superior to TD as consolidation therapy [14]. A study that investigates the effect of Daratumumab in consolidation is also ongoing [15]. Cassiopeia study showed that 2 cycles of Dara-VTD consolidation treatment had a positive effect on PFS in patients with NDMM [10].

Advertisement

5. Treatment of transplant-ineligible patients

Several factors determine the choice of treatment in TI patients. Some authors consider the age limit as 65 years. However, some patients above the age of 65 can have a very good organ function. Therefore, age alone should not be considered the sole determinant for transplantation. Charlson comorbidity index, geriatric assessment scale, and hematopoietic comorbidity index can be used to determine the intensity of treatment. Melphalan is the first agent used in the treatment of elderly myeloma patients. Melphalan and prednisolone (MP) combination can be added to thalidomide (MPT) or bortezomib (VMP). PFS varies between 14 and 62 months among the studies involving MPT [16, 17, 18, 19]. PFS rate is 24 months in Vista Study (bortezomib plus MP treatment) [20].

In a meta-analysis comparing VMP versus MPT, it was found that the CR rate was 21% vs. 13%, PFS 32 months vs. 23 months, and overall survival was 79 months vs. 45 months [21]. One of the most important studies is the SWOG S0777 study in which VRD and RD treatments were compared. PFS rate was 41 months in the VRD group while 29 months in the RD group [22]. Therefore, it supports that bortezomib is one of the most important drugs in induction therapy.

The prognosis has improved with the introduction of lenalidomide as first-line therapy. With the addition of daratumumab to MPV (ALCYON study) or lenalidomide-dexamethasone treatments (MAIA study) as a first-line regimen, the success rate has increased significantly [23, 24]. Today, proteasome inhibitor-IMID-dexamethasone plus a monoclonal antibody combination seems to be the most successful treatment in TI patients. However, it should be emphasized that this treatment is an expensive approach. The results of the studies regarding transplantation in TI patients are presented in Table 4.

Studyn≥ CR
%
PFS
(m)
OS (m)Author
MPT vs. MP850 vs.
772
4–16 vs.
1–8.8
20.3 vs.
14.9
39 vs.
32
Fayers, Blood 2011
VMP vs. MP
(VISTA)
344
338
30 vs. 424 vs. 1656 vs. 43San Miguel, NEJM 2008, JCO 2013
VMP vs. VTD (GEM2005)130 vs. 13020 vs. 2832 vs. 2363 vs. 43Mateos Blood 2014
MP vs. MPR
vs. MPR-R
(MM-015)
154 vs. 152
vs. 153
5 vs. 13
vs. 18
12 vs. 15 vs. 31For 3 years 66% vs. 62% vs. 70%Palumbo, NEJM 2012
RdC vs. RD 18
MPT 12
(FIRST)
535 vs. 541 vs. 54715 vs. 14 vs. 925.5 vs. 20.7 vs. 21.259 vs. 56 vs. 51Benboubker, N Eng J Med 2014
VRD vs. RD
(SWOG S0777)
216 vs. 21415 vs. 841 vs. 29Not reached vs. 69Durie, Lancet 2015 and Blood Cancer Journal 2020
KMP vs. VMP
(CLARION)
478 vs. 47725.9 vs. 23.122.3 vs. 22.1Not reached in all groupFacon, Blood 2019
IRD vs. RD
Tourmaline 2
73 vs. 7425.6 vs. 14.135.3 vs. 21.8Not reachedFacon, Blood 2021
VTD vs. TDxSCT
(Gimema-MMY-3006)
241 vs. 23960 vs. 41110 months
60% vs. 46%
Tahetti, Lancet Hematol, 2020
Dara-LenDex vs. Lendex
(MAIA)
346 vs. 35447.6% vs. 24.9Not reached
31.9
Not reached in all groupsFacon, N Eng J Med 2019
Dara-MPV vs. MPV
(ALCYON)
350 vs. 35642% vs. 24%For 36 months 50% vs. 18.5%Not reached vs. 46 monthsMateos, N Eng J Med 2020

Table 4.

Treatment regimens for TE patients.

m: month (s), n: number, vs.: versus.

Advertisement

6. Maintenance treatment

Maintenance treatment with thalidomide has improved overall survival nonsignificantly. PFS is improved with thalidomide maintenance, but thrombosis risk and peripheral neuropathy incidence are also higher with thalidomide vs. no maintenance. Lenalidomide has also been found a very effective agent for maintenance. PFS and OS were improved in First Study. PFS for Rd. Continue, Rd18, and MPT groups were 16.0, 21, and 21.9 months, respectively. The median OS was found similar in both Rd. Continue, and Rd18 groups (59.1 months vs. 62.3 months) while 49.1 months in the MPT group [25]. Lenalidomide maintenance results are demonstrated in Table 5. The STAMINA study debated the dose and duration of lenalidomide use. In this study, better results were reported in patients who received 15 mg daily lenalidomide treatment continuously [26]. Huang et al. investigated the effect of lenalidomide versus bortezomib maintenance treatment after ASCT. They showed that there is no difference in both arms although adverse effects in the bortezomib arm were higher than in the lenalidomide arm [27].

StudynRegimenPFS
(m)
OS (m)Author
MM0015 IFM153 vs. 154Lenalidomide vs. no Lenalidomide31%77 for 3 yearsPalumbo N Eng J Med 2012
ECOG231 vs. 229Lenalidomide vs. no Lenalidomide46 vs. 27McCharty
Myeloma XI1137 vs. 834Lenalidomide vs. no Lenalidomide39 vs. 2087% vs. 74% for 3 yearsJackson Lancet Oncol 2019
Tourmaline MM3395 vs. 261Ixazomib vs. Placebo26.5 vs. 21.3Dimopoulos Lancet 2019
HOVON213 vs. 219VAD vs. PAD28 vs. 35Sonneveld J Clin Oncol 2012

Table 5.

Maintenance treatment regimens in patients with NDMM.

m: month(s), n: number, vs.: versus.

References

  1. 1. Dispenzieri A, Lacy MQ, Kumar S. Multiple Myeloma. In: Greer JP, editor. Wintrobe’s Clinical Hematology. Under the section: Incidence and Epidemiology. 14th ed. 2019. p. 6411
  2. 2. Cengiz A, Ustun A, Doger F, Yavasoglu F, Yurekli Y, Bolaman AZ. Correlation between baseline 18F-FDG PET/CT findings and CD38- and CD138-expressing myeloma cells in bone marrow and clinical parameters in patients with multiple myeloma. Turkish Journal of Hematology. 2018;35:175-180
  3. 3. Cavo M, Rajkumar SV, Palumbo A, Moreau P, Orlowski R, Bladé J, et al. International myeloma working group consensus approach to the treatment of multiple myeloma patients who are candidates for autologous stem cell transplantation. Blood. 2011;117(23):6063-6073
  4. 4. Larocca A, Palumbo A. How I treat fragile myeloma patient. Blood. 2015;126:2179-2185
  5. 5. Attal M, Harousseau JL, Stoppa AM, Sotto JJ, Fuzibet JG, Rossi JF, et al. A prospective, randomized trial of autologous bone marrow transplantation and chemotherapy in multiple myeloma. Intergroupe Francais du Myelome. The New England Journal of Medicine. 1996;335:91-97
  6. 6. Cavo M, Hájek R, Pantani L, Beksac M, Oliva S, Dozza L, et al. ASCT 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
  7. 7. Moreau P, Hulin C, Macro M, Caillot D, Chaleteix C, Rousselet M, et al. VTD is superior to VCD prior to intensive therapy in multiple myeloma: Results of the prospective IFM2013-04 trial. Blood. 2016;127:2569-2574
  8. 8. Rosiñol L, Oriol A, Rios R, Sureda A, Blanchard MJ, Hernández MT, et al. Bortezomib, lenalidomide, and dexamethasone as induction therapy prior to autologous transplant in multiple myeloma. Blood. 2019;134(16):1337-1345
  9. 9. Kumar S, Jacobus SJ, Cohen AD, Weiss M, Callander NS, Singh AA, et al. Carfilzomib, lenalidomide, and dexamethasone (KRd) versus bortezomib, lenalidomide, and dexamethasone (VRd) for initial therapy of newly diagnosed multiple myeloma (NDMM): Results of ENDURANCE (E1A11) phase III trial. Journal of Clinical Oncology. 2020;38(Suppl. 18) Published online June 01. Available at: https://ascopubs.org/doi/abs/10.1200/JCO.2020.38.18_suppl.LBA3. [Last access date: 01st July, 2022]
  10. 10. Moreau P, Attal M, Hulin C, Arnulf B, Belhadj K, Benboubker L, 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(10192):29-38
  11. 11. Voorhees PM, Rodriguez C, Reeves B, Nathwani N, Costa LJ, Lutska Y, et al. Daratumumab plus RVd for newly diagnosed multiple myeloma: Final analysis of the safety run-in cohort of GRIFFIN. Blood Advances. 2021;5(4):1092-1096
  12. 12. Roussel M, Hebraud B, Lauwers-Cances V, Macro M, Leleu X, Hulin C, et al. Bortezomib and high-dose melphalan vs. high-dose melphalan as conditioning regimen before autologous stem cell transplantation in de novo multiple myeloma patients: A phase 3 study of the Intergroupe francophone Du Myelome (IFM 2014-02). Blood. 2017;130(suppl. 1):398
  13. 13. Straka C, Knop S, Vogel M, Müller J, Kropff M, Metzner B, et al. Bortezomib consolidation following autologous transplant in younger and older patients with newly diagnosed multiple myeloma in two phase III trials. European Journal of Haematology. 2019;103(3):255-267
  14. 14. Cavo M, Pantani L, Petrucci TM, Patriarca, Zamagni E, Donarumma D, et al. GIMEMA (Gruppo Italiano Malattie Ematologiche dell’Adulto) Italian myeloma network Bortezomib-thalidomide-dexamethasone is superior to thalidomide-dexamethasone as consolidation therapy after autologous hematopoietic stem cell transplantation in patients with newly diagnosed multiple myeloma. Blood. 2012;120(1):9-19
  15. 15. Moreau P, Hulin C, Perrot A, Arnulf B, Belhadj K, Benboubker L, et al. Daratumumab maintenance vs observation in patients with newly diagnosed multiple myeloma treated with bortezomib, thalidomide, and dexamethasone ± daratumumab and asct: CASSIOPEIA part 2 results. In: 2021 European Society of Hematology Congress; June 9-17, 2021. Virtual. Abstract S180. https://bit.ly/3pI5QHG. [Accessed: June 11, 2021]
  16. 16. Palumbo A, Bringhen S, Caravita T, Merla E, Capparella V, Callea V, et al. Oral melphalan and prednisone chemotherapy plus thalidomide compared with melphalan and prednisone alone in elderly patients with multiple myeloma: Randomised controlled trial. Lancet. 2006;367(9513):825-831
  17. 17. Facon T, Mary JV, Hulin C, Benboubker L, Attal M, Pegourie B, et al. Melphalan and prednisone plus thalidomide versus melphalan and prednisone alone or reduced-intensity autologous stem cell transplantation in elderly patients with multiple myeloma (IFM 99-06): A randomised trial. Lancet. 2007;370(9594):1209-1218
  18. 18. Hulin C, Facon T, Rodon P, Pegourie B, Benboubker L, Doyen C, et al. Efficacy of melphalan and prednisone plus thalidomide in patients older than 75 years with newly diagnosed multiple myeloma: IFM 01/01 trial. Journal of Clinical Oncology. 2009;27(22):3664-3670
  19. 19. Beksac M, Haznedar R, Tuglular TF, Ozdogu H, Aydogdu İ, Konuk N, et al. Addition of thalidomide to oral melphalan/prednisone in patients with multiple myeloma not eligible for transplantation: Results of a randomized trial from the Turkish myeloma study group. European Journal of Haematology. 2011;86(1):16-22
  20. 20. San Miguel JF, Schlag R, Khuageva NK, Dimopoulos MA, Shpilberg O, Kropff M, et al. Persistent overall survival benefit and No increased risk of second malignancies with Bortezomib-Melphalan-prednisone versus Melphalan-prednisone in patients with previously untreated multiple myeloma. Journal of Clinical Oncology. 2013;31(4):448-455
  21. 21. Morabito F, Bringhen S, Larocca A, Wijermans P, Mateos MV, Gimsing P, et al. Bortezomib, melphalan, prednisone (VMP) versus melphalan, prednisone, thalidomide (MPT) in elderly newly diagnosed multiple myeloma patients: A retrospective case-matched study. American Journal of Hematology. 2014;89(4):355-362
  22. 22. Durie BGM, Hoering A, Sexton R, Abidi MH, Epstein J, Rajkumar SV, et al. Longer term follow-up of the randomized phase III trial SWOG S0777: Bortezomib, lenalidomide and dexamethasone vs. lenalidomide and dexamethasone in patients (pts) with previously untreated multiple myeloma without an intent for immediate autologous stem cell transplant (ASCT). Blood Cancer Journal. 2020;10(5):53
  23. 23. Mateos MV, Dimopoulos MA, Cavo M, Suzuki K, Jakubowiak A, Knop S, et al. Daratumumab plus Bortezomib, Melphalan, and prednisone for untreated myeloma. The New England Journal of Medicine. 2018;378(6):518-528
  24. 24. Facon T, Kumar S, Plesner T, Orlowski RZ, Moreau P, Bahlis N, et al. Daratumumab plus Lenalidomide and dexamethasone for untreated myeloma. The New England Journal of Medicine. 2019;380(22):2104-2115
  25. 25. Benboubker L, Dimopoulos MA, Dispenzieri A, Catalano J, Belch AR, et al. Lenalidomide and dexamethasone in transplant-ineligible patients with myeloma. The New England Journal of Medicine. 2014;371:906-917
  26. 26. Hari P, Pasquini MC, Stadtmauer EA, Fraser R, Fei M, Devine SM, 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). Journal of Clinical Oncology. 2020;38(Suppl. 15):8506-8506
  27. 27. Huang J, Phillips S, Byrne M, Chinratanalab W, Engelhardt BG, Goodman SA, et al. Lenalidomide vs bortezomib maintenance choice post-autologous hematopoietic cell transplantation for multiple myeloma. Bone Marrow Transplantation. 2018;53:701-707

Written By

Ali Zahit Bolaman and Atakan Turgutkaya

Submitted: 27 May 2022 Reviewed: 08 June 2022 Published: 02 July 2022